A Trial Comparing the Efficacy and Safety of Liraglutide 1.8 mg/Day to Liraglutide 0.9 mg/Day in Japanese Subjects With Type 2 Diabetes Mellitus.

July 31, 2018 updated by: Novo Nordisk A/S

A Trial Comparing the Efficacy and Safety of Liraglutide 1.8 mg/Day to Liraglutide 0.9 mg/Day in Japanese Subjects With Type 2 Diabetes Mellitus

This trial is conducted in Asia. The aim of the trial is to compare the efficacy and safety of liraglutide 1.8 mg/day to liraglutide 0.9 mg/day in Japanese subjects with type 2 diabetes mellitus.

Study Overview

Status

Completed

Intervention / Treatment

Study Type

Interventional

Enrollment (Actual)

635

Phase

  • Phase 3

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Locations

      • Annaka-shi, Gunma, Japan, 379 0116
        • Novo Nordisk Investigational Site
      • Chitose, Hokkaido, Japan, 066-0032
        • Novo Nordisk Investigational Site
      • Chuo-ku Tokyo, Japan, 103-0027
        • Novo Nordisk Investigational Site
      • Chuo-ku,, Japan, 104 0061
        • Novo Nordisk Investigational Site
      • Chuo-ku, Tokyo, Japan, 103 0027
        • Novo Nordisk Investigational Site
      • Chuo-ku, Tokyo, Japan, 103 0002
        • Novo Nordisk Investigational Site
      • Fukuoka-shi, Fukuoka, Japan
        • Novo Nordisk Investigational Site
      • Higashiosaka-shi, Osaka, Japan
        • Novo Nordisk Investigational Site
      • Hokkaido, Japan, 078-8236
        • Novo Nordisk Investigational Site
      • Ichikawa-shi, Chiba, Japan
        • Novo Nordisk Investigational Site
      • Iruma-shi, Saitama, Japan, 358 0011
        • Novo Nordisk Investigational Site
      • Izumisano-shi, Japan, 598 0048
        • Novo Nordisk Investigational Site
      • Kashiwara-shi, Osaka, Japan, 582 0005
        • Novo Nordisk Investigational Site
      • Kawagoe-shi, Saitama, Japan, 350 0851
        • Novo Nordisk Investigational Site
      • Kobe-shi, Hyogo, Japan
        • Novo Nordisk Investigational Site
      • Kumamoto-shi,Kumamoto, Japan, 862 0976
        • Novo Nordisk Investigational Site
      • Mito-shi, Ibaraki, Japan, 310-0845
        • Novo Nordisk Investigational Site
      • Mito-shi, Ibaraki, Japan
        • Novo Nordisk Investigational Site
      • Miyazaki-shi, Japan, 880 0034
        • Novo Nordisk Investigational Site
      • Naka-shi, Ibaraki, Japan, 311 0113
        • Novo Nordisk Investigational Site
      • Neyagawa-shi, Osaka, Japan
        • Novo Nordisk Investigational Site
      • Nishinomiya-shi, Hygo, Japan, 662 0971
        • Novo Nordisk Investigational Site
      • Nishinomiya-shi, Hyogo, Japan, 663-8501
        • Novo Nordisk Investigational Site
      • Oita-shi, Japan, 870 0039
        • Novo Nordisk Investigational Site
      • Okawa-shi, Fukuoka, Japan, 831 0016
        • Novo Nordisk Investigational Site
      • Osaka-shi, Osaka, Japan, 553 0003
        • Novo Nordisk Investigational Site
      • Osaka-shi, Osaka, Japan, 5590012
        • Novo Nordisk Investigational Site
      • Oyama-shi, Tochigi, Japan, 323 0022
        • Novo Nordisk Investigational Site
      • Saga-shi,Saga, Japan, 849 0937
        • Novo Nordisk Investigational Site
      • Sapporo-shi, Hokkaido, Japan, 060 0062
        • Novo Nordisk Investigational Site
      • Sapporo-shi, Hokkaido, Japan, 060-0001
        • Novo Nordisk Investigational Site
      • Sapporo-shi, Hokkaido, Japan, 062 0007
        • Novo Nordisk Investigational Site
      • Sappro-shi, Hokkaido, Japan, 060 8648
        • Novo Nordisk Investigational Site
      • Shimotsuke-shi, Tochigi, Japan, 329 0433
        • Novo Nordisk Investigational Site
      • Shinjuku-ku, Tokyo, Japan, 160-0008
        • Novo Nordisk Investigational Site
      • Shizuoka-shi, Japan, 424 0853
        • Novo Nordisk Investigational Site
      • Suita-shi, Osaka, Japan, 565-0853
        • Novo Nordisk Investigational Site
      • Takatsuki-shi, Osaka, Japan, 569 1096
        • Novo Nordisk Investigational Site
      • Tokyo, Japan, 181-0013
        • Novo Nordisk Investigational Site
      • Tokyo, Japan, 103-0028
        • Novo Nordisk Investigational Site
      • Tokyo, Japan, 169-0073
        • Novo Nordisk Investigational Site
      • Tokyo, Japan, 105-8471
        • Novo Nordisk Investigational Site
      • Tokyo, Japan, 123-0845
        • Novo Nordisk Investigational Site
      • Tokyo, Japan, 144-0051
        • Novo Nordisk Investigational Site
      • Yokohama, Kanagawa, Japan, 236-0004
        • Novo Nordisk Investigational Site
      • Yokohama-shi, Japan, 235 0045
        • Novo Nordisk Investigational Site
      • Yokohama-shi Kanagawa, Japan, 232-0064
        • Novo Nordisk Investigational Site

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

20 years and older (Adult, Older Adult)

Accepts Healthy Volunteers

No

Genders Eligible for Study

All

Description

Inclusion Criteria:

  • Male or female Japanese subjects at least 20 years of age at the time of informed consent
  • Type 2 diabetes subjects (diagnosed clinically) for at least 6 months prior to screening
  • HbA1c 7.5-10.0% [58 mmol/mol-86 mmol/mol] (both inclusive)
  • Subjects on stable therapy with one OAD (oral antidiabetic drug) (stable therapy is defined as unchanged medication and unchanged dose) for for at least 60 days before screening according to approved Japanese labelling

Exclusion Criteria:

  • Treatment with insulin within 12 weeks prior to screening
  • Treatment with any medication for the indication of diabetes or obesity other than stated in the inclusion criteria in a period of 60 days before screening
  • Screening calcitonin equal or above 50 ng/l
  • History of pancreatitis (acute or chronic)
  • Personal or family history of medullary thyroid carcinoma (MTC) or multiple endocrine neoplasia type 2 (MEN 2)
  • Subjects presently classified as being in New York Heart Association (NYHA) Class IV
  • Within the past 180 days any of the following: myocardial infarction, stroke or hospitalisation for unstable angina and/or transient ischemic attack
  • Diagnosis of malignant neoplasms within the last 5 years (except basal and squamous cell skin cancer, polyps and in-situ carcinomas)
  • Any condition which, in the opinion of the investigator might jeopardise subject's safety or compliance with the protocol

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: Randomized
  • Interventional Model: Parallel Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: Liraglutide 1.8 mg
The total trial duration for the 1.8 mg/day treatment arm will be approximately 67 weeks, consisting of 2 weeks screening period, a 12 weeks run-in period, a 26-week main treatment period, a safety extension period of 26 weeks and a follow-up visit.
Injected subcutaneously s.c. (under the skin) once daily.
Active Comparator: Liraglutide 0.9 mg
The total trial duration for the 0.9 mg/day treatment arm will be approximately 41 weeks, consisting of 2 weeks screening period, a 12 weeks run-in period, a 26-week treatment period, and a follow-up visit.
Injected subcutaneously s.c. (under the skin) once daily.

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Change in Glycosylated Haemoglobin (HbA1c) (Week 26)
Time Frame: Week 0, Week 26
Change from baseline (week 0) in glycosylated haemoglobin (HbA1c) was evaluated after 26 weeks of treatment. The change from baseline in the response after 26 weeks of treatment is analysed using an analysis of covariance (ANCOVA) model with treatment as a fixed effect and baseline response as a covariate.
Week 0, Week 26

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Change in HbA1c (Week 52)
Time Frame: Week 0, Week 52
Change from baseline (week 0) in HbA1c was evaluated after 52 weeks of treatment.
Week 0, Week 52
Responder for HbA1c Below 7.0% (53 mmol/Mol)
Time Frame: Week 26 and Week 52
Reported results are number of subjects who achieved HbA1c target below 7.0% after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Responder for HbA1c Below or Equal to 6.5% (48 mmol/Mol)
Time Frame: Week 26 and Week 52
Reported results are number of subjects who achieved HbA1c target below or equal to 6.5% after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Responder for HbA1c Below 7.0% Without Weight Gain
Time Frame: Week 26 and Week 52
Reported results are number of subjects who achieved HbA1c target below 7.0% without weight gain after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Responder for HbA1c Below 7.0% Without Treatment Emergent Severe or Blood Glucose (BG) Confirmed Symptomatic Hypoglycaemic Episodes
Time Frame: Week 26 and Week 52
Reported results are subjects with HbA1c <7.0% after 26 weeks and 52 weeks of treatment, respectively without treatment emergent severe or BG confirmed symptomatic hypoglycaemic episodes. Severe or BG confirmed symptomatic hypoglycaemia: severe as per ADA classification or BG confirmed by plasma glucose (PG) value <3.1 mmol/L with symptoms consistent with hypoglycaemia. Severe hypoglycaemia as per ADA: episode requiring assistance of another person to actively administer carbohydrate/glucagon, or take other corrective actions. PG levels may not be available during an event, but neurological recovery following the return of PG to normal is considered sufficient evidence that the event was induced by a low PG level. Treatment emergent: episode with onset date on or after randomisation (from week (wk)0) and no later than 7 days after the last day on liraglutide (maximum till wk26+7days and wk52+7days). Hence, the following shown 'Time Frame' should be read as 'Wk26+7days and Wk52+7days'
Week 26 and Week 52
Change in Self-Measured Blood Glucose (SMBG) 7-point Profile: 7-point Profile (Individual Points in the Profile)
Time Frame: Week 0 and Week 26 and Week 52

Reported results are 7-point SMBG values at week 0, week 26 and week 52. The 7-point profile blood glucose levels were measured at the following time points always starting with the first:

  1. Before breakfast.
  2. 90 minutes after start of breakfast.
  3. Before lunch.
  4. 90 minutes after start of lunch.
  5. Before dinner.
  6. 90 minutes after start of dinner.
  7. At bedtime.
Week 0 and Week 26 and Week 52
Change in SMBG 7-point Profile: Mean of 7-point Profile
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in mean of the SMBG 7-point profile was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in SMBG 7-point Profile: Mean of Postprandial Increments (From Before Meal to 90 Minutes After for Breakfast, Lunch and Dinner)
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in mean of postprandial increments (from before meal to 90 minutes after for breakfast, lunch and dinner) of the SMBG 7-point profile was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Fasting Plasma Glucose (FPG)
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in FPG was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Waist Circumference
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in waist circumference was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Body Weight
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in body weight was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Body Mass Index (BMI)
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in BMI was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Blood Pressure (Systolic and Diastolic)
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in systolic blood pressure (SBP) and diastolic blood pressure (DBP) was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Fasting C-peptide
Time Frame: Week 26 and Week 52
Fasting C-peptide was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Fasting Insulin
Time Frame: Week 26 and Week 52
Fasting insulin was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Fasting Glucagon
Time Frame: Week 26 and Week 52
Fasting glucagon was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Proinsulin
Time Frame: Week 26 and Week 52
Proinsulin was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Proinsulin/Insulin
Time Frame: Week 26 and Week 52
Proinsulin/insulin was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Homeostasis Model Assessment of Beta-cell Function (HOMA-B)
Time Frame: Week 26 and Week 52
HOMA-B was evaluated after 26 weeks and 52 weeks of treatment, respectively. HOMA-B is an index of beta-cell function and was calculated as: HOMA-B=[(20 x fasting insulin in µU/mL)/(FPG in mmol/L-3.5)].
Week 26 and Week 52
Homeostasis Model Assessment as an Index of Insulin Resistance (HOMA-IR)
Time Frame: Week 26 and Week 52
HOMA-IR was evaluated after 26 weeks and 52 weeks of treatment, respectively. HOMA-IR is an index of insulin resistance and was calculated as: HOMA-IR= fasting insulin (μU/mL) x FPG (mmol/L)/22.5.
Week 26 and Week 52
Total Cholesterol
Time Frame: Week 26 and Week 52
Total cholesterol was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Low Density Lipoprotein (LDL) Cholesterol
Time Frame: Week 26 and Week 52
LDL was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
High Density Lipoprotein (HDL) Cholesterol
Time Frame: Week 26 and Week 52
HDL was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Very Low Density Lipoprotein (VLDL) Cholesterol
Time Frame: Week 26 and Week 52
VLDL was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Triglycerides
Time Frame: Week 26 and Week 52
Triglycerides were evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Free Fatty Acids
Time Frame: Week 26 and Week 52
Free fatty acids were evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 26 and Week 52
Number of Treatment Emergent Adverse Events
Time Frame: Weeks 0-26 and Weeks 0-52
Treatment emergent adverse events (TEAEs) were evaluated during the 26-week and 52-week treatment period, respectively. TEAE for weeks 0-26: Event that has onset date on or after randomisation (from week 0) and no later than seven days after the last day on liraglutide (maximum till week 26 + 7 days). TEAE for weeks 0-52: Event that has onset date on or after randomisation (from week 0) and no later than seven days after the last day on liraglutide (maximum till week 52 + 7 days). Hence, the following shown 'Time Frame' should be read as 'Weeks 0-26 + 7 days and Weeks 0-52 + 7 days'.
Weeks 0-26 and Weeks 0-52
Number of Treatment Emergent Severe or BG Confirmed Symptomatic Hypoglycaemic Episodes
Time Frame: Weeks 0-26 and Weeks 0-52
Treatment emergent severe or BG confirmed symptomatic hypoglycaemic episodes were evaluated during the 26-week and 52-week treatment period, respectively. Severe or BG confirmed symptomatic hypoglycaemia (hypo):An episode that was severe according to the ADA classification or BG confirmed by a PG value <3.1 mmol/L with symptoms consistent with hypo. ADA definition of severe hypo:episode requiring assistance of another person to actively administer carbohydrate/glucagon, or take other corrective actions. PG levels may not be available during an event, but neurological recovery following the return of PG to normal is considered sufficient evidence that the event was induced by a low PG level. Treatment emergent: episode with onset date on or after randomisation (from week (wk) 0) and no later than 7 days after the last day on liraglutide (maximum till wk 26 and wk 52, respectively + 7 days). Hence, the following shown 'Time Frame' should be read as 'wk 0-26+7 days and wk 0-52+7 days'.
Weeks 0-26 and Weeks 0-52
Number of Treatment Emergent Nocturnal Severe or BG Confirmed Symptomatic Hypoglycaemic Episodes
Time Frame: Weeks 0-26 and Weeks 0-52
Treatment emergent nocturnal severe or BG confirmed symptomatic hypoglycaemic episodes were evaluated during the26-week and 52-week treatment period, respectively. Nocturnal hypoglycaemic episodes: Those occurring between 00:01 and 05:59 hours, both inclusive. Severe or BG confirmed symptomatic hypoglycaemia: episode that was severe according to the ADA classification or BG confirmed by a PG value <3.1 mmol/L with symptoms consistent with hypoglycaemia. Treatment emergent: episode with onset date on or after randomisation (from week 0) and no later than 7 days after the last day on liraglutide (maximum till week 26 and week 52, respectively + 7 days). Hence, the following shown 'Time Frame' should be read as 'Week 0-26 + 7 days and Week 0-52 + 7 days'.
Weeks 0-26 and Weeks 0-52
Number of Treatment Emergent Hypoglycaemic Episodes According to ADA Definition
Time Frame: Weeks 0-26 and Weeks 0-52

American Diabetes Association (ADA) classification of hypoglycaemia:

  1. Severe: Requiring assistance of another person to actively administer carbohydrate/glucagon/take other corrective actions. PG levels may not be available during an event, but neurological recovery following return of PG to normal is considered sufficient evidence that event was induced by a low PG level.
  2. Documented symptomatic: PG level ≤3.9 mmol/L with symptoms.
  3. Asymptomatic: PG level ≤3.9 mmol/L without symptoms.
  4. Probable symptomatic: No measurement with symptoms.
  5. Pseudo: PG level >3.9 mmol/L with symptoms. Treatment emergent hypoglycaemic episode: episode with onset date on or after randomisation (from week 0) and no later than 7 days after the last day on liraglutide (maximum till week 26 and week 52, respectively + 7 days). Hence, the following shown 'Time Frame' should be read as 'Week 0-26 + 7 days and Week 0-52 + 7 days'.
Weeks 0-26 and Weeks 0-52
Change in Pulse
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in pulse was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Physical Examination
Time Frame: Week 0 and Week 26 and Week 52

Reported results are physical examination outcomes at week (wk) 0, wk 26 and wk 52. Physical examination consisted of the following listed examinations and the outcome of each examination was evaluated as: 1) normal, 2) abnormal, not clinically significant (NCS) or 3) abnormal, clinically significant (CS).

  1. Cardiovascular system
  2. Central and peripheral nervous system (PNS)
  3. Gastrointestinal (GI) system including mouth
  4. General appearance
  5. Head, ears, eyes, nose, throat, neck
  6. Lymph node palpation
  7. Musculoskeletal system
  8. Respiratory system
  9. Skin
  10. Thyroid gland
Week 0 and Week 26 and Week 52
Change in Eye Examination
Time Frame: Week 0 and Week 26 and Week 52
Reported results are eye examination (ophthalmoscopy) outcomes at week 0, week 26 and week 52. Ophthalmoscopy outcomes for both left and right eye were evaluated as: 1) normal, 2) abnormal, NCS or 3) abnormal, CS.
Week 0 and Week 26 and Week 52
Change in Electrocardiogram (ECG)
Time Frame: Week 0 and Week 26 and Week 52
Reported results are ECG outcomes at week 0, week 26 and week 52. ECG outcomes were evaluated as: 1) normal, 2) abnormal, NCS or 3) abnormal, CS.
Week 0 and Week 26 and Week 52
Change in Biochemistry: Creatinine
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in creatinine was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: eGFR
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in estimated glomerular filtration rate (eGFR) was evaluated after 26 weeks and 52 weeks of treatment, respectively. eGFR was evaluated using the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) formula, mL/min/1.73m^2.
Week 0, Week 26, Week 52
Change in Biochemistry: Alanine Aminotransferase
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in alanine aminotransferase was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: Aspartate Aminotransferase
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in aspartate aminotransferase was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: Alkaline Phosphatase
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in alkaline phosphatase was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: Sodium
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in sodium was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: Potassium
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in potassium was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: Albumin
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in albumin was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: Total Bilirubin
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in total bilirubin was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: Urea
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in urea was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: Creatine Kinase
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in creatine kinase was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: Calcium
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in calcium was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: Albumin Corrected Calcium
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in albumin corrected calcium was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: Amylase
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in amylase was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Biochemistry: Lipase
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in lipase was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Haematology: Haemoglobin
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in haemoglobin was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26, Week 52
Change in Haematology: Haematocrit
Time Frame: Week 0, Week 26, Week 52
Change from baseline (week 0) in haematocrit was evaluated after 26 weeks and 52 weeks of treatment, respectively. Haematocrit is the ratio of the volume of red blood cells to the total volume of blood.
Week 0, Week 26, Week 52
Change in Haematology: Thrombocytes
Time Frame: Week 0, Week 26 and Week 52
Change from baseline (week 0) in thrombocytes (platelets) was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26 and Week 52
Change in Haematology: Erythrocytes
Time Frame: Week 0, Week 26 and Week 52
Change from baseline (week 0) in erythrocytes was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26 and Week 52
Change in Haematology: Leukocytes
Time Frame: Week 0, Week 26 and Week 52
Change from baseline (week 0) in leukocytes was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26 and Week 52
Change in Haematology: Eosinophils
Time Frame: Week 0, Week 26 and Week 52
Change from baseline (week 0) in eosinophils was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26 and Week 52
Change in Haematology: Neutrophils
Time Frame: Week 0, Week 26 and Week 52
Change from baseline (week 0) in neutrophils was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26 and Week 52
Change in Haematology: Basophils
Time Frame: Week 0, Week 26 and Week 52
Change from baseline (week 0) in basophils was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26 and Week 52
Change in Haematology: Monocytes
Time Frame: Week 0, Week 26 and Week 52
Change from baseline (week 0) in monocytes was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26 and Week 52
Change in Haematology: Lymphocytes
Time Frame: Week 0, Week 26 and Week 52
Change from baseline (week 0) in lymphocytes was evaluated after 26 weeks and 52 weeks of treatment, respectively.
Week 0, Week 26 and Week 52
Change in Calcitonin
Time Frame: Week 0 and Week 26 and Week 52
Reported results are number of subjects with low, normal or high calcitonin values at week 0, week 26 and week 52. Number of subjects analyzed = number of subjects contributed to the analysis for individual time point. Calcitonin values were categorised as low, normal or high.
Week 0 and Week 26 and Week 52

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (Actual)

July 21, 2015

Primary Completion (Actual)

May 2, 2017

Study Completion (Actual)

November 9, 2017

Study Registration Dates

First Submitted

July 21, 2015

First Submitted That Met QC Criteria

July 21, 2015

First Posted (Estimate)

July 22, 2015

Study Record Updates

Last Update Posted (Actual)

September 5, 2018

Last Update Submitted That Met QC Criteria

July 31, 2018

Last Verified

July 1, 2018

More Information

Terms related to this study

Other Study ID Numbers

  • NN2211-4174
  • U1111-1164-5462 (Other Identifier: WHO)
  • JapicCTI-152975 (Registry Identifier: JAPIC)

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

YES

IPD Plan Description

According to the Novo Nordisk disclosure commitment on novonordisk-trials.com

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

Yes

Studies a U.S. FDA-regulated device product

No

product manufactured in and exported from the U.S.

Yes

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Diabetes Mellitus, Type 2

Clinical Trials on liraglutide

3
Subscribe