throbber
original article
`
`Diabetes, Obesity and Metabolism 16: 748–756, 2014.
`© 2014 John Wiley & Sons Ltd
`
`Dose-finding results in an adaptive, seamless, randomized
`trial of once-weekly dulaglutide combined with metformin
`in type 2 diabetes patients (AWARD-5)
`Z. Skrivanek1, B. L. Gaydos1, J. Y. Chien1, M. J. Geiger2, M. A. Heathman1, S. Berry3, J. H. Anderson4,
`T. Forst5, Z. Milicevic1 & D. Berry3
`1LillyDiabetes,EliLillyandCompany,Indianapolis,IN,USA
`2Cardiovascular&MetabolismTherapeutics,RegeneronPharmaceuticalsInc,Tarrytown,NY,USA
`3BerryConsultants,Austin,TX,USA
`4DiabetesandCardiometabolicMedicine,Carmel,IN,USA
`5Profil,Hellersbergstrasse,Neuss,Germany
`
`article
`original
`
`Aims: AWARD-5 was an adaptive, seamless, double-blind study comparing dulaglutide, a once-weekly glucagon-like peptide-1 (GLP-1)
`receptor agonist, with placebo at 26 weeks and sitagliptin up to 104 weeks. The study also included a dose-finding portion whose results are
`presented here.
`Methods: Type 2 diabetes (T2D) patients on metformin were randomized 3 : 1 : 1 to seven dulaglutide doses, sitagliptin (100 mg), or placebo.
`A Bayesian algorithm was used for randomization and dose selection. Patients were adaptively randomized to dulaglutide doses using available
`data on the basis of a clinical utility index (CUI) of glycosylated haemoglobin A1c (HbA1c) versus sitagliptin at 52 weeks and weight, pulse
`rate (PR) and diastolic blood pressure (DBP) versus placebo at 26 weeks. The algorithm randomly assigned patients until two doses were
`selected.
`Results: Dulaglutide 1.5 mg was determined to be the optimal dose. Dulaglutide 0.75 mg met criteria for the second dose. Dulaglutide 1.5 mg
`showed the greatest Bayesian mean change from baseline (95% credible interval) in HbA1c versus sitagliptin at 52 weeks −0.63 (−0.98 to
`−0.20)%. Dulaglutide 2.0 mg showed the greatest placebo-adjusted mean change in weight [−1.99 (−2.88 to −1.20) kg] and in PR [0.78
`(-2.10 to 3.80) bpm]. Dulaglutide 1.5 mg showed the greatest placebo-adjusted mean change in DBP [−0.62 (−3.40 to 2.30) mmHg].
`Conclusions: The Bayesian algorithm allowed for an efficient exploration of a large number of doses and selected dulaglutide doses of 1.5
`and 0.75 mg for further investigation in this trial.
`Keywords: AWARD-5, Bayesian adaptive, dose finding, dulaglutide dose, GLP-1, GLP-1 receptor agonist, metformin, type 2 diabetes
`
`Date submitted 24 February 2014; date of first decision 12 March 2014; date of final acceptance 18 April 2014
`
`Introduction
`
`Dulaglutide is a long-acting human GLP-1 receptor agonist in
`development as a once-weekly subcutaneous injection for the
`treatment of type 2 diabetes (T2D) [1–3]. The molecule consists
`of two identical, disulphide-linked chains, each containing an
`N-terminal GLP-1 analogue sequence covalently linked to
`a modified human immunoglobulin G4 heavy chain by a
`small peptide linker [1]. Dulaglutide exhibits GLP-1-mediated
`effects, including glucose-dependent potentiation of insulin
`secretion, inhibition of glucagon secretion, delay of gastric
`emptying and weight loss [1–4].
`Dose selection for the dulaglutide clinical development
`programme utilized an adaptive design within the first confir-
`matory dulaglutide trial (AWARD-5) that enabled exploration
`
`Correspondenceto: Zachary Skrivanek, PhD, Eli Lilly Corporate Center, Lilly Diabetes,
`Indianapolis, IN 46285, USA.
`e-mail: skrivanek_zachary@lilly.com
`
`of seven doses in a dose-finding portion, and possible selection
`of up to two doses. The primary and secondary objectives
`compared the efficacy and safety of selected dulaglutide dose(s)
`with sitagliptin at 52 and 104 weeks, and with placebo at
`26 weeks [5,6]. The results of dose-finding are presented here.
`
`Research Design and Methods
`Eligible patients (18–75 years) had T2D (≥6 months) and
`glycosylated haemoglobin A1c (HbA1c) of >8.0% if on
`diet and exercise alone, or HbA1c of ≥7.0 and ≤9.5% if
`on oral antihyperglycaemic medications (OAM) (any OAM
`monotherapy, or combination of metformin with another
`OAM) and a BMI between 25 and 40 kg/m2. The protocol
`was approved by local ethical review boards, and all patients
`provided written informed consent before trial-related activity.
`The study was conducted in compliance with the Declaration of
`Helsinki and the International Conference on Harmonization
`guideline on good clinical practices [7].
`
`MPI EXHIBIT 1113 PAGE 1
`
`

`

`DIABETES, OBESITY AND METABOLISM
`
`AWARD-5 was a multicentre, randomized, double-blind,
`104-week, parallel-arm trial
`in T2D patients treated with
`metformin, and included an initial dose-finding portion
`(Figure 1). The study design and statistical methodologies
`were previously published [8–10]. Eligible patients entered
`a lead-in period (≤11 weeks). Patients were required to be
`treated with metformin (≥1500 mg/day) for ≥6 weeks prior to
`randomization; other OAMs were discontinued. After lead-in,
`patients were randomized to dulaglutide injection (seven doses
`during dose-finding; only selected dose(s) after dose selection
`occurred), sitagliptin 100 mg once daily or placebo (injectable
`and oral), all in combination with metformin.
`The objective of the dose-finding portion was to identify a
`dulaglutide dose with an optimal efficacy and safety profile,
`and possibly one lower dose to be available in case of an
`observed, unforeseen safety signal with the optimal dose
`during subsequent clinical development. For that purpose, a
`Bayesian adaptive algorithm was used. The algorithm involved
`adaptive randomization of patients to dulaglutide doses and
`evaluation of predefined dose decision rules on a biweekly
`basis. The decisions made by the algorithm were based on the
`posterior probability distributions available for each analysis.
`Posterior probability distributions changed with each analysis
`based on the additional data that had accrued in the interim.
`Patients were adaptively randomized to 1 of seven once-weekly
`dulaglutide doses (0.25, 0.50, 0.75, 1.0, 1.5, 2.0 and 3.0 mg),
`preferentially assigning higher probabilities to doses considered
`to have a more favourable clinical profile, and randomization
`to sitagliptin or placebo at 20% each; 3 : 1 : 1 ratio, respectively
`(Figure 1).
`Four efficacy and safety measures were considered important
`for dose selection based on early phase dulaglutide data: HbA1c,
`weight, pulse rate (PR) and diastolic blood pressure (DBP) [1].
`These measures were used to define criteria for dose selection.
`
`original article
`The selected dulaglutide dose(s) had to have a mean change
`of ≤+5 beats per minute (bpm) for PR and ≤+2 mmHg for
`DBP relative to placebo at 26 weeks. In addition, if a dose was
`weight neutral versus placebo, it had to show HbA1c reduction
`≥1.0% and/or be superior to sitagliptin at 52 weeks. If a dose
`reduced weight relative to placebo≥2.5 kg, then non-inferiority
`to sitagliptin would be acceptable.
`A clinical utility index (CUI) was incorporated in the
`algorithm to facilitate adaptive randomization and dose
`selection [8,9] based on the same parameters used to define
`dose-selection criteria described above. Longitudinal models
`were utilized to estimate HbA1c at 52 weeks and weight, PR and
`DBP at 26 weeks. The CUI was applied to estimate for HbA1c,
`weight, DBP and PR. A posterior probability distribution for
`the CUI and its components were calculated every 2 weeks. The
`CUI was a multiplicative index with a range of possible values
`from 0 to 6, with larger values reflecting a favourable clinical
`profile [9]. Zero values resulted if at least one component
`reached prespecified thresholds for a clinically unacceptable
`outcome (i.e. posterior mean increase in DBP ≥2.5 mmHg).
`Decision rules were based on posterior probability thresholds
`and predictive probabilities for meeting the primary study
`objective.
`During each biweekly interim assessment to support the
`adaptive algorithm,
`the dose with the highest posterior
`probability of having the largest CUI was designated the
`maximum utility dose (MUD). After 200 patients were
`randomized (the minimum sample size required for dose
`selection), if the MUD met predefined selection criteria (CUI
`≥0.6 and predictive probability of non-inferiority versus
`sitagliptin at 52 weeks for HbA1c change from baseline ≥0.85)
`at one of the interim assessments, that dose and possibly a
`lower dose would be selected. The lower dose was the next
`highest dose with an acceptable CUI ≥0.6 and ≤50% the dose
`
`Figure 1. Study design. aMetformin concomitant therapy from lead-in through treatment period (≥1500 mg/day). bLead-in period lasted up to 11 weeks.
`cThe dose finding period (indicated by the blue area) ended at the decision point (29 April 2009) resulting in different exposures within and across
`treatment groups. dAfter 26 weeks, patients in the placebo arm transitioned to sitagliptin in a blinded fashion.
`
`Volume 16 No. 8 August 2014
`
`doi:10.1111/dom.12305 749
`
`
`
`MPI EXHIBIT 1113 PAGE 2
`
`

`

`original article
`
`strength of the MUD to ensure minimal overlap of dulaglutide
`exposure. If
`there was strong evidence that no optimal
`dose existed (i.e. no therapeutic window), the algorithm
`would determine the study should be stopped because of
`‘futility’.
`The potential decision point was also adaptive (could have
`happened any time after 200 patients were randomized and
`up until 400 patients were randomized) (Figure 1). If data at
`the decision point supported selection of 1 or 2 doses, patients
`randomized to those specific doses and the comparator arms
`would continue the study, whereas patients assigned to non-
`selected dulaglutide doses would be discontinued. Additional
`patients would be randomized to selected doses and comparator
`arms using a fixed-allocation scheme to test the primary
`objective. The primary objective was to show non-inferiority
`(margin of 0.25%) of the selected optimal dulaglutide dose
`to sitagliptin in HbA1c change from baseline at 52 weeks
`[11]. Patients followed the same visit schedule and testing
`procedures, irrespective of when they were randomized into
`the trial
`An independent Data Monitoring Committee (DMC)
`external to Lilly provided oversight of the implementation of
`the adaptive algorithm and monitored study safety. The DMC
`fulfilled this role during the dose-finding portion, and contin-
`ued monitoring after dose selection until an interim database
`lock at 52 weeks, at which time the study was unblinded to
`assess the primary objectives. Sites and patients continued to
`be blinded to the treatment allocation until the completion
`of the study. The DMC was not allowed to intervene with the
`design operations. A Lilly Internal Review Committee (IRC),
`independent of the study team, would meet if the DMC recom-
`mended the study to be modified. The role of the IRC was to
`make the final decision regarding the DMC’s recommendation.
`The external Statistical Analysis Center (SAC) performed all
`interim data analyses for the DMC, evaluated the decision
`rules and provided the randomization updates for the adaptive
`algorithm.
`The DMC reviewed additional efficacy measures includ-
`ing fasting serum glucose and fasting plasma insulin; β-cell
`function and insulin sensitivity indices [updated Homeosta-
`sis Model Assessment (HOMA2)]; and lipids. Safety assess-
`ments included vital signs, adverse events, laboratory param-
`eters, hypoglycaemic episodes, electrocardiograms (ECGs)
`and dulaglutide antidrug antibodies. Pancreatic enzyme
`measurements began approximately 7 months after study ini-
`tiation, per regulatory guidance for GLP-1 receptor agonists.
`
`Statistical Analysis
`Interim analyses included all patients randomized and were
`based on all available data in the clinical trial database at
`the time of data transfers. After initiation of the adaptive
`algorithm, updated randomization probabilities were provided
`in a biweekly report by the SAC. The report summarized the
`exposure and the randomization probabilities for the next
`2 weeks. Raw data and Bayesian estimates (posterior means,
`posterior probabilities and posterior predictive probabilities)
`for the four CUI components, the CUI and Bayesian parameters
`related to the algorithm were plotted and tabulated. Posterior
`
`DIABETES, OBESITY AND METABOLISM
`
`probability distributions at an interim analysis represent the
`current knowledge about the parameter of
`interest. This
`Bayesian approach is referred to as ‘active learning’, which
`updates posterior distributions with data from the interim
`to help with decision making [9]. Ninety-five percentage
`credible intervals were calculated by taking the 2.5th and
`97.5th percentiles of the corresponding posterior probability
`distributions. The DMC chair and the lead SAC statistician
`reviewed these reports and were tasked to convene an
`unscheduled DMC meeting if an issue was identified with
`the algorithm or the decision point was triggered.
`Additional
`interim safety reports were generated for
`DMC meetings. These reports included summary tables
`of adverse events, serious adverse events, vital signs, ECG
`parameters, hypoglycaemic events, laboratory parameters and
`antihypertensive medications.
`After final database lock at 104 weeks, data from patients
`randomized in the dose-finding portion were reassessed for
`robustness of
`the dose-finding results (Appendix S1 for
`methods, Supporting information and Table S2 for results)
`Statistical analyses were performed using the sas system®
`version 8.2 or higher and Fortran 77.
`
`Results
`Patient Disposition and Exposure
`Patients were adaptively randomized to the seven dulaglutide
`doses during the dose-finding portion (Table S1). A total of
`230 patients were enrolled prior to the decision point. Of
`these, 199 patients had post-randomization data available to
`contribute to the evaluation of the decision rules. The other
`31 patients enrolled shortly before the decision point, and
`thus, had no available post-randomization data. The number
`of patients randomized to treatment arms and their baseline
`characteristics are provided in Table 1. Sponsor decision was the
`most common reason for early study discontinuation (Figure
`S1); this included all patients from dulaglutide arms who were
`discontinued at decision point because their respective doses
`were not selected.
`(10th biweekly interim analysis),
`At decision point
`mean exposure in all dulaglutide-treated patients was
`11.1 ± 7.5 weeks; placebo- and sitagliptin- treated patients had
`been exposed to study drug for (mean± SD) 9.2 ± 7.2 and
`10.7 ± 7.6 weeks, respectively (Table 2).
`
`Dose Selection
`The results of the first nine interim analyses were used only
`for adjustment of the dulaglutide randomization probabilities
`as specified in the protocol. After the ninth interim, the DMC
`recommended ceasing randomization to dulaglutide 3.0 mg
`because of safety concerns (additional data presented below);
`the IRC endorsed that recommendation. The decision rules
`were applied for the first time at the 10th interim, after 200
`patients had been enrolled. Results of the posterior unadjusted
`mean changes from baseline and mean changes from baseline
`adjusted for comparators for the four components of the CUI at
`this interim are shown in Tables 2 and 3, respectively. Figure 2
`
`750 Skrivanek et al.
`
`Volume 16 No. 8 August 2014
`
`
`
`MPI EXHIBIT 1113 PAGE 3
`
`

`

`DIABETES, OBESITY AND METABOLISM
`
`Table 1. Baseline characteristics, demographics and disposition of randomized patients.
`
`original article
`
`Variable
`
`Sex, n (%)
`Men
`Women
`Age (years)
`Race, n (%)
`Black
`White
`East Asian
`Hispanic
`Others
`BMI (kg/m2)
`Duration of
`diabetes (years)
`HbA1c
`[%, (mmol/mol)]
`SBP (mm Hg)
`DBP (mm Hg)
`
`Dulaglutide
`0.25 mg
`(n = 24)
`
`Dulaglutide
`0.5 mg
`(n = 25)
`
`Dulaglutide
`0.75 mg
`(n = 21)
`
`Dulaglutide
`1.0 mg
`(n= 10)
`
`Dulaglutide
`1.5 mg
`(n = 25)
`
`Dulaglutide
`2.0 mg
`(n = 30)
`
`Dulaglutide
`3.0 mg
`(n = 15)
`
`Sitagliptin
`(n = 42)
`
`Placebo
`(n= 38)
`
`9 (38)
`15 (63)
`57 (7)
`
`0 (0)
`12 (50)
`5 (21)
`6 (25)
`1 (4)
`31 (4)
`7 (4)
`
`7.8 (0.8)
`[62 (9)]
`130 (15)
`78 (9)
`
`13 (52)
`12 (48)
`55 (10)
`
`2 (8)
`9 (36)
`0 (0)
`14 (56)
`0 (0)
`33 (5)
`7 (4)
`
`10 (48)
`11 (52)
`52 (11)
`
`0 (0)
`13 (62)
`1 (5)
`7 (33)
`0 (0)
`33 (5)
`7 (5)
`
`3 (30)
`7 (70)
`55 (9)
`
`0 (0)
`4 (40)
`0 (0)
`6 (60)
`0 (0)
`34 (4)
`7 (5)
`
`10 (40)
`15 (60)
`53 (11)
`
`2 (8)
`10 (40)
`2 (8)
`11 (44)
`0 (0)
`32 (5)
`9 (7)
`
`8 (27)
`22 (73)
`53(11)
`
`3 (10)
`13 (43)
`6 (20)
`8 (27)
`0 (0)
`31 (5)
`7 (5)
`
`5 (33)
`10 (67)
`53 (11)
`
`1 (7)
`7 (47)
`0 (0)
`7 (47)
`0 (0)
`31 (5)
`7 (6)
`
`21 (50)
`21 (50)
`53 (12)
`
`2 (5)
`20 (48)
`4 (10)
`16 (38)
`0 (0)
`32 (4)
`9 (5)
`
`12 (32)
`26 (68)
`53 (10)
`
`2 (5)
`15 (40)
`4 (11)
`17 (45)
`0 (0)
`32 (4)
`7 (6)
`
`8.3 (1.3)
`[67 (14)]
`125 (16)
`76 (7)
`
`8.2 (1.1)
`[66 (12)]
`133 (16)
`82 (8)
`
`7.9 (0.6)
`[63 (7)]
`134 (13)
`79 (10)
`
`8.7 (1.5)
`[72 (16)]
`127 (16)
`77 (8)
`
`8.4 (1.0)
`[68 (11)]
`127 (15)
`79 (8)
`
`8.0 (1.1)
`[64 (12)]
`128 (17)
`76 (9)
`
`8.4 (1.1)
`[68 (12)]
`128 (12)
`77 (6)
`
`8.1 (1.1)
`[65 (12)]
`127 (12)
`78 (8)
`
`Data are means (SD) or n (%) unless otherwise indicated. BMI, body mass index; DBP, diastolic blood pressure; HbA1c, glycosylated haemoglobin A1C;
`SBP, systolic blood pressure; SD, standard deviation.
`
`provides the Bayesian estimates for the CUI and for each
`individual component. All results presented are based on data
`accrued up until the time of dose selection, the decision point.
`Posterior mean (95% credible interval) changes from
`baseline in HbA1c at 52 weeks ranged from 0.82 (−1.13,
`−0.53)% for dulaglutide 0.25 mg to−1.33 (−1.62,−1.00)% for
`dulaglutide 1.5 mg (Table 2 and Figure 2). The posterior mean
`change for sitagliptin was −0.70 (−0.87, −0.56)%. Dulaglutide
`1.5 mg showed the greatest posterior mean change compared
`to sitagliptin at 52 weeks −0.63 (−0.98, −0.20)% (Table 3).
`Posterior mean (95% credible interval) changes from
`baseline in weight at 26 weeks ranged from −0.96 (−1.47,
`−0.33) kg for dulaglutide 0.75 mg to −4.45 (−5.30, −3.70) kg
`for dulaglutide 3.0 mg (Table 2 and Figure 2). The posterior
`mean change from baseline for placebo was −0.52 (−0.99,
`0.01) kg (Table 2 and Figure 2). Across the range of dulaglutide
`doses (excluding discontinued 3.0 mg dose), dulaglutide 2.0 mg
`showed the greatest posterior mean change in weight compared
`to placebo at 26 weeks [−1.99 (−2.88, −1.20) kg] (Table 3).
`For PR, the posterior mean (95% credible interval) changes
`from baseline to 26 weeks for dulaglutide doses ranged from
`1.47 (−1.50, 4.13) bpm for dulaglutide 0.5 mg to 6.26 (3.95,
`10.95) bpm for dulaglutide 3.0 mg (Table 2 and Figure 2). The
`posterior mean change for placebo was 3.27 (1.23, 5.47) bpm
`(Table 2 and Figure 2). Across the range of dulaglutide
`doses (excluding discontinued 3.0 mg dose), dulaglutide 2.0 mg
`showed the greatest posterior mean change in PR compared to
`placebo at 26 weeks 0.78 (−2.10, 3.80) bpm (Table 3).
`The posterior mean (95% credible interval) changes from
`baseline to 26 weeks in DBP for dulaglutide doses ranged
`from −0.02 (−1.74, 1.78) mmHg for dulaglutide 0.25 mg to
`−0.99 (−2.22, 0.20) mmHg for dulaglutide 1.5 mg (Table 2
`and Figure 2). The posterior mean change for placebo was
`−0.37 (−2.88, 1.99) mmHg (Table 2 and Figure 2). Across
`
`the range of dulaglutide doses (excluding discontinued 3.0 mg
`dose), dulaglutide 1.5 mg showed the greatest posterior mean
`change in DBP compared to placebo at 26 weeks [−0.62 (−3.40,
`2.30) mmHg] (Table 3).
`Dulaglutide 1.5 mg was determined to be the MUD [3.1
`(0.7, 4.0)] at the 10th interim assessment (Figure 2). The
`posterior probability that the CUI for this dose of ≥0.6 sample
`size was 0.982 and the posterior predictive probability that
`dulaglutide 1.5 mg would show non-inferiority to sitagliptin
`at 52 weeks, based on a total sample size of 263 in each
`arm, was >0.99 (Figure 2). On the basis of these results, the
`algorithm determined the decision point had been reached, and
`selected two doses: dulaglutide 1.5 mg as the optimal dose and
`dulaglutide 0.75 mg as the second, lower dose (CUI= 1.054).
`It also determined the total sample size for each treatment arm
`in the trial based on the data from the dose-finding portion.
`The population PK/PD results supported the Bayesian-
`based results. A summary of the range of expected responses
`predicted by population PK/PD exposure-response models is
`provided in the Table S2 (Appendix S1 for details in method).
`These model-predicted responses and CUI values supported
`the Bayesian-based results shown in Figure 2. Dulaglutide
`1.5 mg was associated with a similar effect on HbA1c (mean;
`90% confidence interval) −1.27 (−1.72 to −0.84)% and weight
`−3.49 (−5.32 to 2.07) kg to that estimated during the dose-
`finding portion. Dulaglutide 1.5 mg also met the prespecified
`requirements of change in PR ≤5 bpm. No concentration-
`response relationship was identified for DBP, hence, there are
`no results presented for this CUI component.
`Data for all nine treatment arms from the final database up
`to the decision point were summarized for each component
`of the CUI. These reports (data not shown) were consistent
`with the results of assessments based on the datasets used for
`adaptation by the SAC.
`
`Volume 16 No. 8 August 2014
`
`doi:10.1111/dom.12305 751
`
`
`
`MPI EXHIBIT 1113 PAGE 4
`
`

`

`original article
`
`Table 2. Bayesian posterior mean (95% credible interval) changes from baseline and exposure.
`
`DIABETES, OBESITY AND METABOLISM
`
`Dose (mg)
`
`Dulaglutide 0.25
`
`Dulaglutide 0.5
`
`Dulaglutide 0.75
`
`Dulaglutide 1.0
`
`Dulaglutide 1.5
`
`Dulaglutide 2.0
`
`Dulaglutide 3.0
`
`Sitaglitpin
`
`Placebo
`
`HbA1c (%)
`52 weeks
`−0.82
`(−1.13; −0.53)
`n= 13
`−0.95
`(−1.16; −0.75)
`n= 16
`−0.93
`(−1.17; −0.65)
`n= 20
`−1.00
`(−1.25; −0.69)
`n= 8
`−1.33
`(−1.62; −1.00)
`n= 18
`−1.28
`(−1.46; −1.07)
`n= 24
`−1.00
`(−1.30; −0.78)
`n= 10
`−0.70
`(−0.87; −0.56)
`n= 25
`0.01
`(−0.27; 0.27)
`n= 28
`
`Weight (kg)
`26 weeks
`−1.01
`(−1.53; −0.35)
`n = 16
`−1.48
`(−2.04; −0.86)
`n = 22
`−0.96
`(−1.47; −0.33)
`n = 20
`−2.05
`(−2.75; −1.34)
`n = 9
`−2.18
`(−2.68; −1.74)
`n = 22
`−2.50
`(−3.20; −1.78)
`n = 29
`−4.45
`(−5.30; −3.70)
`n = 13
`−0.45
`(−0.89; 0.06)
`n = 35
`−0.52
`(−0.99; 0.01)
`n = 33
`
`Pulse rate (bpm)
`26 weeks
`
`2.12
`(−0.83; 3.84)
`n = 16
`1.95
`(−0.35; 4.15)
`n = 22
`1.47
`(−1.50; 4.13)
`n = 20
`2.56
`(0.19; 4.34)
`n = 9
`3.2
`(1.25; 4.99)
`n = 22
`4.05
`(2.63; 5.70)
`n = 29
`6.26
`(3.95; 10.95)
`n = 13
`−0.90
`(−2.80; 1.09)
`n = 35
`3.27
`1.23; 5.47)
`n = 33
`
`DBP (mm Hg)
`26 weeks
`−0.02
`(−1.74; 1.78)
`n = 16
`−0.26
`(−1.67; 0.81)
`n = 22
`−0.65
`(−1.92; 0.34)
`n = 20
`−0.92
`(−2.36; 0.38)
`n = 9
`−0.99
`(−2.22; 0.20)
`n = 22
`−0.78
`(−2.05; 0.47)
`n = 29
`−0.57
`(−2.70; 1.85)
`n = 13
`−1.22
`(−3.52; 0.75)
`n = 35
`−0.37
`(−2.88; 1.99)
`n = 33
`
`Mean utility
`
`0.733
`(0, 1.639)
`
`1.362
`(0, 2.326)
`
`1.054
`(0.090, 1.923)
`
`2.021
`(0.415, 3.267)
`
`3.052
`(0.721, 4.037)
`
`2.996
`(0, 3.702)
`
`Not applicable
`
`Not applicable
`
`Not applicable
`
`Exposure (weeks)
`mean ± SD
`8.9± 8.5
`
`9.7± 7.1
`
`15.7± 6.2
`
`13.0± 8.8
`
`12.5± 7.4
`
`10.0± 5.2
`
`9.2± .8.8
`
`10.7± 7.6
`
`9.2± 7.2
`
`Statistics presented are posterior means and 95% credible intervals based on data available at the decision point (10th Interim, 29 April 2009). bpm, beats
`per minute; DBP, diastolic blood pressure; HbA1c, glycosylated haemoglobin A1c; SD, standard deviation.
`
`Safety
`The most common adverse events reported during dose-
`finding were gastrointestinal (including nausea, diarrhoea and
`vomiting) and urinary tract infection (Table S3). Nausea and
`vomiting were more common with dulaglutide than sitagliptin
`and placebo. The highest incidence was observed in patients
`with doses ≥1.0 mg. There were a total of three serious adverse
`events (SAEs) (dulaglutide 0.5 mg: pneumonia [1]; dulaglutide
`3.0 mg: gastroenteritis [1] and placebo: cervical dysplasia
`[1]). Approximately 1 year after study discontinuation, a
`patient exposed to dulaglutide 2.0 mg for 6 months was
`diagnosed with an SAE of medullary thyroid carcinoma
`(MTC). The patient had increased calcitonin levels before
`randomization [91.5 pg/ml (reference range 0–11.5 pg/ml),
`evaluated retrospectively] which decreased upon exposure to
`dulaglutide. At the time of dulaglutide discontinuation and
`3 months thereafter, the patient’s calcitonin was 61.7 and
`82.8 pg/ml, respectively. Subsequently, this patient was also
`found to be positive for the RET proto-oncogene mutation,
`indicating a preexisting neoplasm in an individual with high
`risk of MTC. At the time of this patient’s randomization,
`there were no exclusionary criteria for patients at increased
`risk of C-cell neoplasm. Four patients each in the dulaglutide
`2.0 mg, dulaglutide 3.0 mg, and placebo arms, and 1 each in the
`dulaglutide 0.25 mg and dulaglutide 1.0 mg arms, discontinued
`
`the study because of adverse events. Nausea was the most
`commonly reported adverse event that led to discontinuation
`with dulaglutide (2.0 mg: one patient; 3.0 mg: three patients).
`In the placebo group, hyperglycaemia was the most common
`adverse event resulting in discontinuation (two patients).
`The proportion of patients with post-baseline lipase levels
`greater than ×3 upper limit of normal (ULN) was 2.5%
`(dulaglutide 3.0 mg one patient; dulaglutide 2.0 mg two
`patients; sitagliptin one patient). Increase in lipase above
`ULN was the most common treatment-emergent abnormal
`laboratory finding and ranged from 13% for placebo to 46%
`for dulaglutide 2.0 mg. Similar increases in pancreatic amylase
`were reported, but were lower in frequency. There were no
`events of pancreatitis reported during dose-finding.
`Dulaglutide 3.0 mg was discontinued because of increase in
`PR (posterior mean for PR >5 bpm) and higher incidence
`of gastrointestinal adverse events (nausea and vomiting)
`with/without higher increases in pancreatic enzymes compared
`to lower doses.
`
`Discussion
`AWARD-5 was the first Phase 3 trial of dulaglutide. The
`first portion of the trial served as the dose finding and dose
`selection trial for the dulaglutide development programme. An
`algorithm was employed to adaptively randomize patients to
`
`752 Skrivanek et al.
`
`Volume 16 No. 8 August 2014
`
`
`
`MPI EXHIBIT 1113 PAGE 5
`
`

`

`DIABETES, OBESITY AND METABOLISM
`
`Table 3. Bayesian posterior adjusted mean (95% credible interval) changes from baseline.
`
`Dose (mg)
`
`Dulaglutide 0.25
`
`Dulaglutide 0.5
`
`Dulaglutide 0.75
`
`Dulaglutide 1.0
`
`Dulaglutide 1.5
`
`Dulaglutide 2.0
`
`Dulaglutide 3.0
`
`HbA1c (%)
`52-week
`sitagliptin-adjusted
`−0.12
`(−0.38; 0.20)
`n= 13
`−0.24
`(−0.44; 0.02)
`n= 16
`−0.23
`(−0.44; 0.08)
`n= 20
`−0.30
`(−0.58; 0.08)
`n= 8
`−0.63
`(−0.98; −0.20)
`n= 18
`−0.58
`(−0.76; −0.32)
`n= 24
`−0.29
`(−0.62; −0.02)
`n= 10
`
`Weight (kg)
`26-week
`placebo-adjusted
`−0.49
`(−1.36; 0.44)
`n = 16
`−0.97
`(−1.80; −0.12)
`n = 22
`−0.44
`(−1.16; 0.44)
`n = 20
`−1.53
`(−2.44; −0.60)
`n = 9
`−1.67
`(−2.48; −0.92)
`n = 22
`−1.99
`(−2.88; −1.20)
`n = 29
`−3.93
`(−4.84; −2.88)
`n = 13
`
`Pulse rate (bpm)
`26-week
`placebo-adjusted
`−1.16
`(−4.40; 1.90)
`n = 16
`−1.32
`(−4.30; 1.80)
`n = 22
`−1.80
`(−5.50; 2.10)
`n = 20
`−0.71
`(−4.00; 2.50)
`n = 9
`−0.07
`(−3.20; 2.70)
`n = 22
`0.78
`(−2.10; 3.80)
`n = 29
`2.98
`(−0.60; 8.70)
`n = 13
`
`original article
`
`DBP (mm Hg)
`26-week
`placebo-adjusted
`
`0.35
`(−2.60; 3.60)
`n = 16
`0.11
`(−2.40; 3.20)
`n = 22
`−0.27
`(−3.00; 2.50)
`n = 20
`−0.55
`(−3.90; 2.20)
`n = 9
`−0.62
`(−3.40; 2.30)
`n = 22
`−0.41
`(−2.60; 3.20)
`n = 29
`−0.19
`(−2.70; 2.80)
`n = 13
`
`Mean utility
`
`0.733
`(0, 1.639)
`
`1.362
`(0, 2.326)
`
`1.054
`(0.090, 1.923)
`
`2.021
`(0.414, 3.267)
`
`3.052
`(0.721, 4.037)
`
`2.996
`(0, 3.702)
`
`Not applicable
`
`Statistics presented are model predicted means and 95% credible intervals based data available at the decision point. bpm, beats per minute; DBP, diastolic
`blood pressure; HbA1c, glycosylated haemoglobin A1c.
`
`a range of seven dulaglutide doses and to select up to two
`doses. This design enabled simultaneous assessment of efficacy
`and safety measures deemed relevant for dose selection based
`on the results of the completed clinical pharmacology studies,
`optimized patient allocation using data accumulated during
`the dose-finding portion and selected the optimal dose when
`enough data to reach a decision were available. Dulaglutide
`1.5 mg was selected as the optimal dose and dulaglutide 0.75 mg
`was selected as the lower dose for further assessment.
`The data met the protocol prespecified conditions for
`dose selection and the assignment algorithm dropped all but
`two active doses, dulaglutide 1.5 and 0.75 mg, at the 10th
`interim analysis. Dulaglutide 1.5 mg had the highest posterior
`probability of being the MUD. Assessment of the effects of
`dulaglutide 1.5 mg on the posterior probability distributions
`for the four components of the CUI showed that this dose had
`robust glucose and weight lowering effects, and was associated
`with an increase in PR that was smaller than the effect of the
`adjacent 2.0 mg dose. These observations explain the selection
`of the 1.5 mg dose. The 0.75 mg dose met the prespecified
`criteria for a lower dulaglutide dose. The selection of the
`dulaglutide 1.5 and 0.75 mg doses were also supported by
`PK/PD model-based analyses and the results of the efficacy and
`safety assessments up to 104 weeks [5,6,12–16].
`The dose responses for PR and weight are different than for
`HbA1c. It appears, based on this study (Table 2) and previous
`studies [1], that the dose-response attenuates around 1.5 mg
`for HbA1c but continues for weight and PR. This closely linked
`dose effect for weight loss is in line with expectations for the
`GLP-1 receptor agonist class and may have significant value
`
`in combination with other antidiabetes therapies that cause
`weight gain (e.g. insulin).
`Although dulaglutide was commonly associated with GI
`effects in earlier Phase 1 studies, that effect was not considered
`to be dose limiting in the dose range included in the AWARD-5
`study. Phase 1 data indicated that PR and DBP would be dose
`limiting before GI would become a factor in dose selection.
`Because there was no indication of potential dose–response
`effect for SBP, PR and DBP were included in the CUI. In this
`study we did not observe any dose response with DBP, but PR
`was indeed dose limiting. There were no DMC concerns related
`to GI issues with the selected doses.
`the DMC
`After
`the ninth interim data assessment,
`recommended discontinuing the dulaglutide 3.0 mg dose
`because of increases in PR, higher incidence of GI adverse
`events, and higher incidence in pancreatic enzyme values
`above ULN. These changes were not unexpected based on the
`known dose-response effects of the GLP-1 receptor agonist
`class, and observations from completed dulaglutide trials
`[1,12,17,18]. Although the changes in pancreatic enzymes have
`not been completely understood, recent reports have showed
`an increased incidence of asymptomatic increases above upper
`limit of the normal range in lipase and amylase in patients with
`T2D [19–23]. The incidence increases further with exposure
`to GLP-1 receptor agonists; these asymptomatic findings have
`not been associated with any adverse outcome in clinical trials
`[24,25].
`The Bayesian adaptive approach used in AWARD-5 has
`not been used often in drug development, although it offers
`substantial benefits in terms of accuracy and efficiency in
`
`Volume 16 No. 8 August 2014
`
`doi:10.1111/dom.12305 753
`
`
`
`MPI EXHIBIT 1113 PAGE 6
`
`

`

`original article
`
`DIABETES, OBESITY AND METABOLISM
`
`Figure 2. Dose–response model and CUI. CUI and change from baseline in CUI components, posterior means and 95% credible intervals at 6 months
`(DBP, PR and weight) and 12 months (HbA1c) (data available at decision point, the 10th Interim, 29 April 2009). bpm, beats per minute; CUI, clinical
`utility index; DBP, diastolic blood pressure; DU, dulaglutide; HbA1c, glycosylated haemoglobin A1C; PK/PD, pharmacokinetic/pharmacodynamics; PL,
`placebo; PR, pulse rate; SITA, sitagliptin.
`
`comparison to a fixed design [26,27]. Several design aspects [use
`of prior information (information gained from Phase 1 studies,
`the mechanism of action and general knowledge about the
`class), adaptive randomization and dose selection] were utilized
`to increase the efficiency and quality of dose-finding. First, the
`Bayesian approach explicitly used prior information to inform
`the dose response and the longitudinal profiles of the treatment
`arms. This decreased the uncertainty of the CUI, allowing for
`earlier adaptations and decision making. Second, the adaptive
`randomization enabled exploration of more dulaglutide doses
`than would be possible with a traditional fixed-allocation
`strategy, given total sample size constraints. This was achieved
`by allowing data accumulating during the trial to drive
`randomization probabilities, in effect, shifting patients away
`from doses with a less favourable clinical profile to doses more
`probably to meet target clinical profiles (Figure 2). Finally,
`
`the adaptive algorithm selected doses when it determined
`adequate safety and efficacy information was collected; whereas
`a fixed design would stop after a predetermined study length
`irrespective of whether there was sufficient information to make
`a decision. The efficiency of the utilized design is showed by the
`relatively small sample size (230 patients) and short exposures
`(median exposure: 4–16 weeks) needed

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