throbber
Diabetes & Metabolism 46 (2020) 100–109
`
`Available online at
`
`ScienceDirect
`www.sciencedirect.com
`
`Original article
`
`Efficacy and safety of once-weekly semaglutide 1.0 mg vs once-daily
`liraglutide 1.2 mg as add-on to 1–3 oral antidiabetic drugs in subjects
`with type 2 diabetes (SUSTAIN 10)
`
`M.S. Capehorn a,*, A.-M. Catarig b, J.K. Furberg b, A. Janez c, H.C. Price d, S. Tadayon b,
`B. Verge` s e, M. Marre f
`a Rotherham Institute for Obesity, Clifton Medical Centre, S65 1DA Rotherham, South Yorkshire, UK
`b Novo Nordisk A/S, DK-2860 Søborg, Denmark
`c Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
`d West Hampshire Community Diabetes Service, SO43 7NG Lyndhurst, UK
`e CHU, 21000 Dijon, France
`f Clinique Ambroise Pare´, 27, boulevard Victor-Hugo, 92200 Neuilly-sur-Seine, France
`
`A R T I C L E
`
`I N F O
`
`A B S T R A C T
`
`Article history:
`Received 14 June 2019
`Received in revised form 28 August 2019
`Accepted 1st September 2019
`Available online 17 September 2019
`
`Keywords:
`Glycaemic control
`Glucagon-like peptide-1 receptor agonist
`Liraglutide
`Semaglutide
`Type 2 diabetes
`SUSTAIN
`
`Aims. – SUSTAIN 10 compared the efficacy and safety of the anticipated most frequent semaglutide dose
`(1.0 mg) with the current most frequently prescribed liraglutide dose in Europe (1.2 mg), reflecting
`clinical practice.
`Methods. – In this phase 3b, open-label trial, 577 adults with type 2 diabetes (HbA1c 7.0–11.0%) on 1–3
`oral antidiabetic drugs were randomized 1:1 to subcutaneous once-weekly semaglutide 1.0 mg or
`subcutaneous once-daily liraglutide 1.2 mg. Primary and confirmatory secondary endpoints were
`changes in HbA1c and body weight from baseline to week 30, respectively.
`Results. – Mean HbA1c (baseline 8.2%) decreased by 1.7% with semaglutide and 1.0% with liraglutide
`(estimated treatment difference [ETD] –0.69%; 95% confidence interval [CI] 0.82 to 0.56, P < 0.0001).
`Mean body weight (baseline 96.9 kg) decreased by 5.8 kg with semaglutide and 1.9 kg with liraglutide (ETD
`3.83 kg; 95% CI 4.57 to 3.09, P < 0.0001). The proportions of subjects achieving glycaemic targets
`of < 7.0% and  6.5%, weight loss of  5% and  10%, and a composite endpoint of HbA1c < 7.0% without
`severe or blood glucose-confirmed symptomatic hypoglycaemia and no weight gain were greater with
`semaglutide vs liraglutide (all P < 0.0001). Both treatments had similar safety profiles, except for more
`frequent gastrointestinal disorders (the most common adverse events [AEs]) and AEs leading to premature
`treatment discontinuation with semaglutide vs liraglutide (43.9% vs 38.3% and 11.4% vs 6.6%, respectively).
`Conclusion. – Semaglutide was superior to liraglutide in reducing HbA1c and body weight. Safety profiles
`were generally similar, except for higher rates of gastrointestinal AEs with semaglutide vs liraglutide.
` C 2019 Elsevier Masson SAS. All rights reserved.
`
`Abbreviations: AACE, American Association of Clinical Endocrinologists; ADA, American Diabetes Association; AE, adverse event; BG, blood glucose; BMI, body mass index;
`bpm, beats per minute; CI, confidence interval; CKD-EPI, The Chronic Kidney Disease Epidemiology Collaboration; CoV, coefficient of variation; CV, cardiovascular; DPP-4i,
`dipeptidyl peptidase-4 inhibitor; DTSQs, Diabetes Treatment Satisfaction Questionnaire status version; E, number of events; EASD, European Association for the Study of
`Diabetes; eGFR, estimated glomerular filtration rate; exenatide ER, exenatide extended release; ETD, estimated treatment difference; ETR, estimated treatment ratio; FAS, full
`analysis set; FPG, fasting plasma glucose; GI, gastrointestinal; GLP-1, glucagon-like peptide-1; GLP-1RA, glucagon-like peptide-1 receptor agonist; KDIGO, Kidney Disease
`Improving Global Outcomes; max., maximum; MedDRA, Medical Dictionary for Regulatory Activities; MET, metformin; min., minimum; MTD, maximum tolerated dose; n,
`number of subjects; N, total number of subjects; OAD, oral antidiabetic drug; OD, once daily; OR, odds ratio; OW, once weekly; PRO, patient-reported outcome; R, event rate
`per 100 exposure-years; SAS, safety analysis set; s.c., subcutaneous; SD, standard deviation; SE, standard error; SGLT-2i, sodium–glucose cotransporter-2 inhibitor; SF-36v21,
`Short-Form 36 Health Survey version 21; SMBG, self-measured blood glucose; SU, sulfonylurea; SUSTAIN, Semaglutide Unabated Sustainability in Treatment of Type
`2 Diabetes; TEAE, treatment-emergent adverse event; T2D, type 2 diabetes.
`* Corresponding author. Rotherham Institute for Obesity (RIO), Clifton Medical Centre, The Health Village, Doncaster Gate, Rotherham S65 1DA, UK.
`E-mail address: mcapehorn@yahoo.co.uk (M.S. Capehorn).
`
`https://doi.org/10.1016/j.diabet.2019.101117
`1262-3636/ C 2019 Elsevier Masson SAS. All rights reserved.
`
`Novo Nordisk Exhibit 2082
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00001
`
`

`

`M.S. Capehorn et al. / Diabetes & Metabolism 46 (2020) 100–109
`
`101
`
`Introduction
`
`There are currently a variety of treatment options for type
`2 diabetes (T2D); despite this, a large proportion of subjects do not
`treatment
`targets
`[1]. Furthermore, optimal
`achieve HbA1c
`treatment of T2D should involve patient-oriented treatment goals
`include minimizing
`extending beyond glycaemic control, to
`like weight gain and hypoglycaemia, and
`unwanted effects
`reducing the risk of complications such as cardiovascular (CV)
`events [2,3].
`Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have
`emerged as effective treatments for T2D and are incorporated into
`the clinical guidelines [2,3]. The 2018 American Diabetes Associa-
`tion (ADA)/European Association for the Study of Diabetes (EASD)
`consensus report and the 2019 ADA Standards of Care treatment
`guidelines recommend preferred treatments, following metformin
`(MET), as either GLP-1RAs or sodium–glucose cotransporter-2
`(SGLT-2is), particularly
`in adults with T2D and
`inhibitors
`additional CV
`risk
`factors
`(e.g. GLP-1RAs or SGLT-2is
`for
`established atherosclerotic disease or SGLT-2is for chronic kidney
`disease or heart failure) [2,3]. In addition to improving glycaemic
`control, some drugs in the GLP-1RA class also provide weight loss,
`have CV benefits, improve renal outcomes, and minimize hypo-
`glycaemic risk [2–7]. Several GLP-1RAs are currently available,
`both short- and long-acting, as once-daily (OD) or once-weekly
`(OW) injections [8], with different molecular sizes and structures
`that result in varying efficacy and safety profiles [9,10].
`Semaglutide (Novo Nordisk A/S) is a long-acting glucagon-like
`peptide-1 (GLP-1) analog, approved for the treatment of T2D in a
`subcutaneous (s.c.), OW formulation [11,12]. The efficacy and
`safety of semaglutide OW has been investigated in the Semaglutide
`Unabated Sustainability in Treatment of Type 2 Diabetes (SUS-
`TAIN) phase 3 clinical trial program across the continuum of care in
`subjects with T2D. In the SUSTAIN trials semaglutide consistently
`demonstrated superior reductions in HbA1c and body weight vs
`placebo and a range of active comparators, including other GLP-
`1RAs (exenatide extended release [ER] and dulaglutide) and basal
`insulin glargine, with a safety profile similar to that of other GLP-
`1RAs [4,13–18]. Furthermore, and in line with findings with the
`GLP-1 analog liraglutide in the LEADER trial [5], the SUSTAIN 6 trial
`demonstrated that semaglutide significantly reduced the risk of
`major adverse CV events, compared with placebo, in subjects with
`T2D and high CV risk [4].
`The aim of the SUSTAIN 10 trial (NCT03191396) was to compare
`the efficacy and safety of OW semaglutide 1.0 mg with OD
`liraglutide 1.2 mg in adults with T2D. These doses were chosen to
`reflect clinical practice regarding use of GLP-1RAs in Europe: OW
`is expected to be the most
`frequently
`semaglutide 1.0 mg
`prescribed dose, whereas OD liraglutide 1.2 mg is currently the
`most frequently prescribed dose [19].
`
`Methods
`
`Trial design
`
`SUSTAIN 10 was a 30-week, randomized, multicentre, multi-
`national, active-controlled, parallel-group, open-label, two-armed
`phase 3b trial conducted in 11 European countries (Bulgaria, Czech
`Republic, Finland, France, Hungary, Italy, Poland, Slovenia, Spain,
`Sweden, United Kingdom). The trial design is shown in Figure S1
`(see supplementary materials associated with this article on line)
`and a full list of trial investigators is shown in Table S1 (see
`supplementary materials associated with this article on line). The
`trial was conducted in compliance with the International Council
`on Harmonisation of Technical Requirements for Registration of
`
`Pharmaceuticals for Human Use Good Clinical Practice guidelines
`[20], and the Declaration of Helsinki [21]. Subjects provided
`consent before the commencement of any trial-related activities.
`The protocol is available in the Appendix (see supplementary
`materials associated with this article on line).
`
`Participants
`
`Key inclusion criteria were age  18 years; T2D with HbA1c 7.0–
`11.0%; and stable daily doses of any of the following antidiabetic
`drug(s) or combination regimens 90 days prior to screening:
`biguanides (MET  1500 mg or maximum tolerated dose [MTD]),
`(SU) or SGLT-2i
`(for both SU and SGLT-2i:
`sulfonylurea
` 0.5 maximum approved dose according to local label or MTD
`as documented in subject medical record). Key exclusion criteria
`were renal impairment, measured as estimated glomerular filtration
`rate (eGFR) < 30 mL/min/1.73 m2; presence of New York Heart
`Association Class IV heart failure; proliferative retinopathy or
`maculopathy requiring acute treatment, verified by fundus photog-
`raphy or dilated fundoscopy within the 90 days prior to randomiza-
`tion; impaired liver function (alanine aminotransferase  2.5 times
`upper limit of normal at screening); and presence or history of
`malignant neoplasms within the past 5 years prior to screening. Full
`trial eligibility criteria are listed in Table S2 (see supplementary
`materials associated with this article on line).
`
`Randomization
`
`Subjects with T2D inadequately controlled on 1–3 oral antidiabetic
`drug(s) (OADs) were randomized 1:1 to treatment with either s.c. OW
`semaglutide 1.0 mg or s.c. OD liraglutide 1.2 mg (both supplied by
`Novo Nordisk A/S, Bagsværd, Denmark). Subjects were stratified
`based on background medication of SU and SGLT2-i (SU  MET; SGLT-
`2i  MET; SU and SGLT-2i  MET; MET monotherapy).
`
`Treatments
`
`Following a 2-week screening period, the treatment period was
`30 weeks, with a 5-week safety data collection follow-up to
`accommodate for the long half-life of semaglutide. After randomi-
`followed a dose-escalation regimen. The
`zation, all subjects
`semaglutide maintenance dose was reached after an 8-week
`escalation period consisting of 4 weeks of 0.25 mg OW, followed by
`4 weeks of 0.5 mg OW. The liraglutide maintenance dose was
`reached after 1 week of 0.6 mg OD. In the event of unacceptable
`gastrointestinal (GI) adverse events (AEs) with liraglutide, escala-
`tion from 0.6 mg to 1.2 mg could be extended over 2 weeks at the
`discretion of the investigator. Both medications were administered
`by injections to the thigh, abdomen, or upper arm, at any time of
`irrespective of meals. Semaglutide
`injections were
`day and
`administered OW preferably on the same day, while liraglutide
`injections were administered OD at the same time every day.
`Subjects were intended to continue on stable, pre-trial background
`medication dose(s) throughout treatment, unless rescue criteria
`were met or a safety concern relating to the background
`medication arose.
`Rescue medication (intensification of antidiabetic background
`medication and/or initiation of new antidiabetic medication) was
`if subjects experienced persistent and unacceptable
`offered
`hyperglycaemia (fasting plasma glucose [FPG] levels  13.3 m-
`mmol/L from week 8 to the end of week 15, or  11.1 mmol/L from
`week 16 to the end of treatment). Rescue medication was
`prescribed at the investigators’ discretion according to ADA/EASD
`2012 and 2015 guidelines [22,23]; GLP-1RAs, dipeptidyl pepti-
`inhibitors
`(DPP-4is), and amylin analogs were not
`dase-4
`permitted.
`
`Novo Nordisk Exhibit 2082
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00002
`
`

`

`102
`
`Endpoints
`
`M.S. Capehorn et al. / Diabetes & Metabolism 46 (2020) 100–109
`
`The primary endpoint was change in HbA1c (%-point, hereafter
`referred to as ‘%’) from baseline to week 30. The confirmatory
`secondary endpoint was change in body weight (kg) from baseline
`to week 30. Other pre-specified supportive secondary efficacy
`endpoints included changes from baseline to week 30 in: FPG;
`mean postprandial increment across all meals and mean 7-point
`profile based on self-measured blood glucose (SMBG); fasting
`blood lipids (total cholesterol, low-density lipoprotein-cholesterol,
`lipoprotein-cholesterol, and
`triglycerides); body
`high-density
`mass index (BMI); waist circumference; and systolic and diastolic
`blood pressure.
`included subjects
`Pre-specified clinical treatment targets
`who, after 30 weeks of treatment, achieved HbA1c < 7.0% (ADA)
`[24] or  6.5% (American Association of Clinical Endocrinolo-
`gists) [25]. In addition, the proportions of subjects achieving
`reduction  1%; weight
`loss  3%,  5%, or  10%; a
`HbA1c
`composite endpoint of HbA1c < 7.0% without severe
`(ADA
`classification) [26] or blood glucose (BG)-confirmed symptom-
`atic hypoglycaemic episodes and no weight gain; and composite
`reduction of  1% and weight
`loss
`endpoints of HbA1c
`of  3%,  5%, or  10%. Supportive secondary endpoints
`for
`(PROs)
`included changes
`from
`patient-reported outcomes
`baseline to week 30 in Short-Form 36 Health Survey version
`21 (SF-36v21) and Diabetes Treatment Satisfaction Question-
`naire status version (DTSQs) scores [27,28].
`Safety endpoints included the number of treatment-emergent
`adverse events (TEAEs, classified as events that had an onset date,
`‘on-treatment’ observation
`or increase in severity, during the
`period) and the number of treatment-emergent severe or BG-
`confirmed symptomatic hypoglycaemic episodes. Other safety
`endpoints included change from baseline to week 30 in haema-
`tology, biochemistry, calcitonin, pulse rate, electrocardiogram
`category, physical examination category, and eye examination
`category. All AEs were coded using the most recent version of the
`Medical Dictionary for Regulatory Activities (MedDRA).
`
`Statistical analysis
`
`The primary estimand was defined as the treatment difference
`between semaglutide and liraglutide at week 30 for all randomized
`subjects if all subjects completed treatment and did not initiate
`rescue medication. This estimand was considered clinically
`relevant as it assessed the glycaemic benefit a subject with T2D
`was expected to achieve if they initiated and continued treatment
`with semaglutide vs liraglutide.
`Efficacy endpoints were evaluated based on the full analysis set
`included all randomized subjects)
`from the
`‘on-
`(FAS, which
`treatment without rescue medication’ observation period; safety
`endpoints were analyzed using the safety analysis set (SAS, which
`included data from all subjects exposed to at least one dose of trial
`product) from the ‘on-treatment’ or ‘in-trial’ observation periods. See
`Table S3 and protocol (see supplementary materials associated with
`this article on line) for the definitions of the observation periods.
`Three confirmatory hypotheses were tested using the following
`hierarchical testing procedure [29]:
`
`1 HbA1c non-inferiority of semaglutide 1.0 mg vs
`1.2 mg (non-inferiority margin of 0.3);
`2 Body weight superiority of semaglutide 1.0 mg vs liraglutide
`1.2 mg;
`3 HbA1c superiority of semaglutide 1.0 mg vs liraglutide 1.2 mg
`[Figure S2 (see supplementary materials associated with this
`article on line)].
`
`liraglutide
`
`The Type-I error rate for testing the three confirmatory
`hypotheses relating to HbA1c and body weight was preserved at
`an overall one-sided alpha (a) level of 2.5%. A sample size of
`in each of the semaglutide and
`288 subjects was needed
`liraglutide groups (total planned randomized: 576 subjects), to
`provide at least 90% power to reject all three confirmatory
`hypotheses and, thus, confirm HbA1c superiority and body weight
`superiority of semaglutide vs liraglutide across efficacy and in-
`trial assumptions.
`The primary analysis addressed the primary estimand, which
`was based on the FAS using measurements up to and including
`‘on-treatment without rescue medication’
`week 30 from the
`observation period.
`In the primary analysis, imputation of missing data was
`handled using multiple imputation assuming that missing data
`imputed as
`were missing at random. Missing data were
`intermittent values using a Markov Chain Monte Carlo method
`to obtain a monotone missing data pattern. This imputation was
`done for each treatment group separately and 500 copies of the
`dataset were generated. A sequential regression approach for
`imputing monotonely missing values at planned visits was
`implemented starting with the first visit after baseline and
`sequentially continued to the last planned visit (week 30). A
`linear model was applied to each treatment group. This model
`used the background medication stratification factor (SU  MET,
`SGLT-2i  MET, SU and SGLT-2i  MET, and MET monotherapy) as a
`categorical effect, and baseline and post-baseline HbA1c values
`(observed and imputed) prior to the visit in question as covariates.
`An analysis of covariance with treatment and background medica-
`tion stratification factor as categorical effects and baseline HbA1c as
`a covariate were used to analyze HbA1c at week 30 for each of the
`500 data sets generated as part of the imputation of missing values.
`Rubin’s rule was used to combine the analysis results in order to
`draw inference. Sensitivity analyses (tipping-point, retrieved drop-
`out [superiority only], and per-protocol [non-inferiority only]
`analyses) were conducted on the primary analysis, see Table S3 (see
`supplementary materials associated with this article on line) for
`details.
`The secondary confirmatory endpoint of change from baseline
`to week 30 in body weight was analyzed in the same way as the
`primary endpoint, but using baseline and post-baseline body
`weight measurements as covariates (instead of HbA1c). Sensitivity
`analyses (tipping-point and retrieved drop-out analyses) were also
`conducted on the secondary confirmatory endpoint, see Table S3
`(see supplementary materials associated with this article on line)
`for details.
`Continuous endpoints were analyzed separately using a similar
`model approach as for the primary endpoint, with associated
`baseline values as covariates (instead of HbA1c). The binary
`endpoints were analyzed using a logistic regression model with
`treatment and stratification factor as fixed factors and baseline
`values as covariates. Before analysis, missing data for individual
`components were imputed separately using the same approach as
`for continuous endpoints and subsequently dichotomized. The
`PRO questionnaires (SF-36v21 and DTSQs) were used to evaluate
`life and treatment satisfaction; see Table S3 (see
`quality of
`supplementary materials associated with this article on line) for
`further details on the PRO questionnaires.
`Safety outcomes were summarized descriptively based on
`the SAS using data the from ‘on-treatment’ observation period,
`retinopathy, which were
`except neoplasms and diabetic
`from the
`‘in-trial’ observation period.
`reported using data
`Summaries of treatment-emergent hypoglycaemic episodes
`were presented as an overview, including all episodes and
`episodes by severity.
`
`Novo Nordisk Exhibit 2082
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00003
`
`

`

`M.S. Capehorn et al. / Diabetes & Metabolism 46 (2020) 100–109
`
`103
`
`Results
`
`Between June and November 2017, 767 subjects were screened,
`of whom 577 were randomized and 576 were exposed to
`treatment (Fig. 1). Of the FAS, a total of 287 (99.0%) subjects in
`the OW semaglutide 1.0 mg arm and 282 (98.3%) subjects in the OD
`liraglutide 1.2 mg arm completed the trial; 249 (85.9%) and 261
`(90.9%) completed treatment, respectively.
`Baseline characteristics and background medications were
`generally similar in each treatment group. The overall mean age
`was 59.5 years, HbA1c 8.2%, body weight 96.9 kg, and diabetes
`duration 9.3 years. Most subjects (94.8%) received biguanides;
`46.8% of subjects received SU and 24.6% received SGLT-2i. Few
`subjects in either treatment arm required rescue medication
`(4 subjects with semaglutide vs 12 subjects with liraglutide; all
`in the
`liraglutide group were treatment
`subjects except one
`completers; Table 1; Fig. 1). Diabetes complications at screening
`are shown in Table S4 (see supplementary materials associated
`with this article on line).
`Mean HbA1c (baseline 8.2%) decreased over time for both
`treatment arms (Fig. 2a), and from baseline to week 30 (Fig. 2b) by
`liraglutide (estimated
`1.7% with semaglutide and 1.0% with
`treatment difference (ETD) at week 30 0.69% [95% confidence
`interval (CI) 0.82 to 0.56], P < 0.0001 for superiority). The
`results of the primary analysis were supported by the sensitivity
`analyses.
`FPG was reduced with both semaglutide and liraglutide from
`baseline to week 30, but changes were significantly greater with
`(ETD 1.24 mmol/L
`[95% CI 1.54
`to 0.93],
`semaglutide
`P < 0.0001, Fig. 2c). Observed SMBG 7-point profile (at baseline
`and week 30) and change in mean 7-point SMBG profile from
`baseline to week 30 are shown in Fig. 2d and e, respectively;
`reductions in the mean profile were greater with semaglutide vs
`(ETD 0.89 mmol/L
`[95% CI 1.15
`to 0.64],
`liraglutide
`
`P < 0.0001). Reductions in the SMBG increment from baseline to
`week 30 were also greater with semaglutide vs liraglutide (ETD
`0.53 mmol/L [95% CI 0.77 to 0.28], P < 0.0001; data not
`shown).
`The proportions of subjects achieving HbA1c < 7.0% and  6.5%
`at week 30 were 80% vs 46% and 58% vs 25%, respectively, with
`semaglutide vs liraglutide (estimated odds ratio [OR] 5.98 [95% CI
`3.83 to 9.32] and 4.84 [95% CI 3.21 to 7.30], respectively,
`P < 0.0001; Fig. 2f and g). A greater proportion of subjects achieved
`HbA1c reduction  1% with semaglutide vs liraglutide (83% vs 48%
`respectively, estimated OR 7.24 [95% CI 4.55 to 11.50], P < 0.0001;
`Figure S3a [see supplementary materials associated with this
`article on line]).
`Mean body weight (baseline 96.9 kg) decreased over time for
`both treatment arms (Fig. 3a), and from baseline to week 30
`(Fig. 3b) by 5.8 kg vs 1.9 kg with semaglutide vs liraglutide (ETD
`3.83 kg [95% CI 4.57 to 3.09], P < 0.0001). The results of the
`confirmatory analysis were supported by the sensitivity analyses.
`The proportions of subjects achieving weight loss of  5%
`and  10% at week 30 were 56% vs 18% and 19% vs 4%, respectively,
`with semaglutide vs liraglutide (estimated OR 5.89 [95% CI 3.93 to
`8.81] and 4.99 [95% CI 2.57 to 9.68], respectively, both P < 0.0001;
`Fig. 3c and d). Similarly, a greater proportion of subjects also
`achieved weight loss  3% with semaglutide vs liraglutide (73% vs
`34% respectively, estimated OR 5.22 [95% CI 3.57 to 7.62],
`P < 0.0001); Figure S3b (see supplementary materials associated
`with this article on line).
`Changes in BMI and waist circumference from baseline to week
`30 were also significantly greater with semaglutide than with
`liraglutide [Table S5 (see supplementary materials associated with
`this article on line)].
`The composite endpoint of HbA1c < 7.0% without severe or BG-
`confirmed symptomatic hypoglycaemia and without weight gain
`was achieved by a greater proportion of subjects treated with
`
`Fig. 1. Subject disposition. One subject receiving liraglutide 1.2 mg discontinued treatment prematurely, the primary reason for which was a non-treatment-emergent
`adverse event. *Screened subjects who withdrew consent before randomization; yincludes only exposed subjects; zsubjects who completed the trial according to the end-of-
`trial form; §subjects who completed treatment according to the end-of-treatment form; ôone extra subject in the liraglutide group received rescue medication but did not
`complete treatment.
`
`Novo Nordisk Exhibit 2082
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00004
`
`

`

`104
`
`M.S. Capehorn et al. / Diabetes & Metabolism 46 (2020) 100–109
`
`Table 1
`Baseline characteristics – full analysis set.
`
`Age, years
`Sex, male
`Race, White
`HbA1c, %
`Fasting plasma glucose, mmol/L
`Diabetes duration, years
`Body weight, kg
`Body mass index, kg/m2
`eGFR, mL/min/1.73 m2 geometric mean (CoV)
`Renal function, mL/min/1.73 m2a
`eGFR  90
`eGFR  60– < 90
`eGFR 30– < 60
`Antidiabetes medication at screening
`Biguanides
`Sulfonylurea
`SGLT-2i
`DPP-4ib
`Other blood glucose-lowering drugs, excluding insulinb
`
`Semaglutide
`1.0 mg
`(n = 290)
`
`60.1 (10.5)
`160 (55.2%)
`264 (91.0%)
`8.2 (0.9)
`9.8 (2.3)
`9.6 (6.1)
`96.6 (21.0)
`33.7 (6.6)
`91.3 (20.3)
`
`190 (65.5%)
`86 (29.7%)
`14 (4.8%)
`
`279 (96.2%)
`136 (46.9%)
`73 (25.2%)
`0
`1 (0.3%)
`
`Liraglutide
`1.2 mg
`(n = 287)
`
`58.9 (10.0)
`167 (58.2%)
`268 (93.4%)
`8.3 (1.0)
`9.9 (2.5)
`8.9 (5.7)
`97.2 (21.7)
`33.7 (7.0)
`89.7 (23.1)
`
`185 (64.5%)
`88 (30.7%)
`14 (4.9%)
`
`268 (93.4%)
`134 (46.7%)
`69 (24.0%)
`1 (0.3%)
`0
`
`Total
`(N = 577)
`
`59.5 (10.2)
`327 (56.7%)
`532 (92.2%)
`8.2 (1.0)
`9.9 (2.4)
`9.3 (5.9)
`96.9 (21.3)
`33.7 (6.8)
`90.5 (21.7)
`
`375 (65.0%)
`174 (30.2%)
`28 (4.9%)
`
`547 (94.8%)
`270 (46.8%)
`142 (24.6%)
`1 (0.2%)
`1 (0.2%)
`
`Data are mean (SD) or n (%) for the full analysis set (FAS), unless otherwise stated. Baseline information is defined as the measurement at the latest assessment before dosing.
`Body mass index is calculated based on baseline measurement of body weight and height.
`%: percentage of subjects; CKD-EPI: The Chronic Kidney Disease Epidemiology Collaboration; CoV: coefficient of variation; DPP-4i: dipeptidyl peptidase-4 inhibitor; eGFR:
`estimated glomerular filtration rate; max.: maximum; min.: minimum; n: number of subjects; N: total number of subjects; SD: standard deviation; SGLT-2i: sodium–glucose
`cotransporter-2 inhibitor.
`a The renal function categories are based on the eGFR using CKD-EPI. No subjects had an eGFR < 30 mL/min/1.73 m2.
`b The two subjects on DPP-4is and repaglinide were randomized in error and discontinued treatment.
`
`semaglutide vs liraglutide (76% vs 37% respectively, estimated OR
`6.07 [95% CI 4.02 to 9.15], P < 0.0001; Fig. 4a).
`A greater proportion of subjects treated with semaglutide vs
`also
`achieved
`composite
`endpoints of HbA1c
`liraglutide
`reduction  1% and different weight-loss responses of  3%,
` 5%, and  10% body weight: 62% vs 21%
`(estimated OR
`6.63 [95% CI 4.44 to 9.91], P < 0.0001; Figure S4a [see supplemen-
`tary materials associated with this article on line]), 50% vs 12%
`(estimated OR 7.55 [95% CI 4.80 to 11.88], P < 0.0001; Figure S4b
`[see supplementary materials associated with this article on line]),
`and 17% vs 4% (estimated OR 5.26 [95% CI 2.58 to 10.73],
`P < 0.0001; Fig. 4b), respectively.
`The change from baseline (136.4 mmHg) to week 30 in systolic
`blood pressure was moderate with both semaglutide (4.5 mmHg
`[standard error (SE) 0.7]) and liraglutide (3.5 [0.7]), and the ETD
`between the treatment arms was not significant (1.0 [95% CI 3.0
`to 1.1] Table S5 [see supplementary materials associated with this
`article on line]). Similarly, there was no significant difference in
`diastolic blood pressure between treatments [Table S5 (see
`supplementary materials associated with this article on line)].
`Changes from baseline to week 30 for lipid levels were modest
`for both treatments, but the semaglutide group showed signifi-
`cantly greater improvements vs the liraglutide group for total
`triglycerides
`[Table S6
`(see supplementary
`cholesterol and
`materials associated with this article on line)].
`in PRO scores were
`reported with both
`Improvements
`treatment arms. The DTSQs showed a significant difference
`between treatment groups in ‘Feeling of unacceptably high blood
`favouring
`sugars’, with this aspect of treatment satisfaction
`semaglutide (ETD 0.55 [95% CI 0.83 to 0.27], P = 0.0001;
`Table S7 [see supplementary materials associated with this article
`on line]). The SF-36v21 questionnaire showed significant diffe-
`rences between the two treatment groups in two components of
`health-related quality of life, with the results favouring sema-
`glutide: vitality (ETD 1.68 [95% CI 0.45 to 2.92], P = 0.0076) and
`mental health (ETD 1.30 [95% CI 0.06; 2.53], P = 0.0396; Table S7
`[see supplementary materials associated with this article on
`
`line]). There were no significant differences in any other DTSQs
`scales or SF-36v21 domains
`[Table S7
`(see supplementary
`materials associated with this article on line)].
`In total, 70.6% (n = 204) subjects experienced TEAEs in the
`semaglutide group, and 66.2% (n = 190) in the liraglutide group
`(Table 2). TEAEs were mainly mild to moderate in severity. A
`slightly higher number of subjects experienced serious TEAEs with
`liraglutide (n = 22, 7.7%) than with semaglutide (n = 17, 5.9%).
`in either treatment group. A higher
`There were no deaths
`proportion of subjects reported TEAEs
`leading to premature
`treatment discontinuation with semaglutide (n = 33, 11.4%) vs
`liraglutide (n = 19, 6.6%); this was primarily driven by GI AEs (7.6%
`with semaglutide vs 3.8% with liraglutide).
`The most commonly reported AEs were GI disorders, reported
`in 127 (43.9%) subjects with semaglutide and 110 (38.3%) subjects
`with liraglutide. The onset of GI AEs was typically during the initial
`12 weeks of the trial. GI AEs were most prevalent during the dose-
`escalation period (liraglutide) or within the first 12 weeks of
`treatment (semaglutide); events were generally mild in severity.
`Nausea was the most frequently reported GI AE, reported by 63
`(21.8%) vs 45 (15.7%) subjects with semaglutide vs liraglutide
`(Table 2). Other frequently reported GI AEs with semaglutide and
`liraglutide were: diarrhea (15.6% and 12.2%), vomiting (10.4% and
`8.0%), constipation (5.9% and 3.5%), and abdominal pain (5.2% and
`2.1%). A list of the AEs reported in  5% of subjects in either
`treatment arm is shown in Table S8 (see supplementary materials
`associated with this article on line).
`Severe or BG-confirmed symptomatic hypoglycaemia was
`experienced by 1.7% of subjects (n = 5; 8 events) in the semaglutide
`group and 2.4% of subjects (n = 7; 8 events) in the liraglutide group;
`no subject in either group experienced severe hypoglycaemic
`episodes (ADA definition; data not shown). Of the 16 episodes of
`severe or BG-confirmed symptomatic hypoglycaemia, 15 were in
`subjects receiving background SU.
`Pancreatitis TEAEs (pre-defined MedDRA search) were reported
`in two (0.7%) subjects receiving liraglutide and no subjects receiving
`semaglutide. Neoplasms (benign, malignant, and unspecified) were
`
`Novo Nordisk Exhibit 2082
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00005
`
`

`

`M.S. Capehorn et al. / Diabetes & Metabolism 46 (2020) 100–109
`
`105
`
`Fig. 2. Glycaemic endpoints with semaglutide 1.0 mg and liraglutide 1.2 mg. Estimated change in HbA1c by week (a); estimated change in HbA1c from overall baseline mean to
`week 30 (b); estimated change in FPG over time (c); observed mean 7-point SMBG profile at baseline and at week 30 (d); mean 7-point SMBG profile change from baseline to
`week 30 (e); proportion of subjects achieving HbA1c < 7.0% (f) and  6.5% (g) at week 30. *P < 0.0001 vs liraglutide 1.2 mg. All figures based on the full analysis set, using ‘on-
`treatment without rescue medication’ data. Figures a–c and e: mean estimates are from an analysis of covariance, where missing data were accounted for using multiple
`imputation (data from subjects within the same group defined by randomized treatment) using a regression model including stratification factor as categorical effect and data
`from baseline and all previous post-baseline visits as covariates. Error bars are  standard errors of the means. Dashed grey lines indicate the overall mean values at baseline.
`Values in square brackets are 95% CIs. Figure d: dashed lines indicate baseline values; solid lines indicate week 30 data. SMBG assessed with glucose meter as plasma equivalent
`values of capillary whole blood glucose. Figures f, g: missing HbA1c data were accounted for using multiple imputation (data from subjects within the same group defined by
`randomized treatment) using a regression model including stratification factor as categorical effect and data from baseline and all previous post-baseline visits as covariates. After
`imputation, continuous data were dichotomized. AACE: American Association of Clinical Endocrinologists; ADA: American Diabetes Association; CI: confidence interval; ETD:
`estimated treatment difference; FPG: fasting plasma glucos

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