The Effects of Different Doses of Exercise on Pancreatic β-cell Function in Patients With Newly Diagnosed Type 2 Diabetes (DOSE-EX)

December 12, 2018 updated by: Mathias Ried-Larsen

The Effects of Different Doses of Exercise on Pancreatic ß-cell Function in Patients With Newly Diagnosed Type 2 Diabetes (DOSE-EX): A Randomized Clinical Trial

This project will provide an exercise-based lifestyle intervention with the potential to reduce complications for patients with short standing type 2 diabetes (T2D). While exercise is widely accepted as a component of T2D management, little is known about the additive effect of exercise when combined with a diet on T2D pathophysiology and mechanisms believed to lead to micro- and macrovascular complications. Moreover, the necessary dose of exercise to revert the progression of T2D and the related complications has not been investigated. A large-scale randomized controlled trial (RCT) will be essential to document the effectiveness on reducing the risk of T2D complications. However, prior to conducting a large-scale RCT, we need to specify the exercise dose that efficiently compliments the diet.

In a 4-armed randomized, clinical trial (N=80 T2D patients, T2D duration < than 7 years) we aim to investigate 1) the potential additive role of exercise on pancreatic β-cell function in patients with T2D when combined with a diet, 2) the causal relationship between lifestyle-induced reductions in glycaemic variability, oxidative stress and low-grade inflammation and, 3) the role of exercise in rescuing dysregulated muscle progenitor cells. The participants will be randomly allocated to either a) control, b) diet, c) diet and exercise 3 times/week or d) diet and exercise 6 times/week for 16 weeks. Prior to, during and following the interventions, all participants will undergo extensive testing.

Study Overview

Detailed Description

A 4-armed, 16-week, parallel-group, assessor-blinded, randomized, clinical trial. Participants will be randomly allocated (1:1:1:1), stratified by sex

Interventions:

The lifestyle interventions will consist of two main components; 1) increased physical activity and structured exercise and/or 2) a dietary intervention aiming at a weight loss. Whereas there will be no differences in the dietary intervention between the lifestyle groups, the volume of physical activity and structured exercise will vary according to the frequencies of the structured exercise sessions.

The study groups are prescribed:

  1. Control group (CON): No intervention
  2. Dietary control (DCON): Dietary intervention (see below)
  3. Moderate Exercise Dose (MED): Two aerobic training sessions per week of 45-60 min duration and one session per week with combined aerobic (30-35 min) and resistance (30 min) training and a dietary intervention (described below)
  4. High Exercise Dose (HED): Four aerobic training sessions per week of 45-60 min duration and two sessions per week with combined aerobic (30-35 min) and resistance (30 min) training and a dietary intervention (described below)

Detailed description of the intervention components. Exercise: The training protocol will be adapted based on a previous study where the T2D participants were prescribed 6 weekly sessions of aerobic training alone or combined aerobic and resistance training (averaging 360-420 min of exercise per week). As previous analyses suggest that there may be an inverse dose-response relationship between reductions in HbA1c and aerobic exercise volume, this parameter will be used to adapt the training protocol. As the effect of exercise on HbA1c is closer related to the number of training sessions rather than intensity15, we will reduce the number of sessions by 50%, to three sessions/week in the moderate exercise dose group and maintain the original session frequency in the high dose exercise group (six sessions/week).Training will be supervised and monitored to ensure intensity and compliance.

Dietary intervention and intended weight loss (DCON, MED and HED: The dietary intervention will be based on the recommendations from the American Diabetes Association (ADA) with increased focus on macronutrient quality. The macronutrient distributions are in line with the current guidelines from the national Diabetes Association and Canadian guidelines, where individualization in macronutrient distribution should lie within the range of 45-60 energy% carbohydrate, 15-20 energy% protein and 20-35 energy% fat. Thus, the dietary intervention emphasis will be on low glycemic index and low glycemic load in shape of non-processed foods and will aim at reducing saturated fat intake <7 energy%.

Study Type

Interventional

Enrollment (Anticipated)

80

Phase

  • Not Applicable

Contacts and Locations

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

Study Contact

Study Contact Backup

Study Locations

      • Copenhagen, Denmark, 2100
        • Recruiting
        • Center for Physical Activity Research, Copenhagen University Hospital
        • Contact:
        • Contact:

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

16 years to 78 years (Adult, Older Adult)

Accepts Healthy Volunteers

No

Genders Eligible for Study

All

Description

Inclusion Criteria:

Diagnosed with diabetes type 2 and/or HbA1c ≥ 48 mmol/mol if no treatment with anti-diabetic medication and/or use of antidiabetic medication

Caucasian

No diagnose of Type 1 diabetes, mature onset diabetes of the young, Latent autoimmune diabetes of adults

T2D 0-6 years of duration

No treatment with insulin

Body Mass Index (BMI) >27 kg/m2 and <40 kg/m2

No known or signs of intermediate or severe microvascular complications to diabetes (retino-, neuro- or nephropathy)

No known cancer

No Known lung disease

No known cardiovascular disease

No known thyroid disease

No known liver disease

No known autoimmune disease

No other endocrine disorder causing obesity

No current treatment with anti-obesity medication

No current treatment with anti-inflammatory medication

No weight loss of > 5kg within the last 6 months

No diagnose of depression or treatment with anti-depressive medication, ongoing or within the last three months before enrolment

No diagnose of psychiatric disorder or treatment with anti-psychotic medication

No history of suicidal behavior or ideations within the last three months before enrolment

No previous surgical treatment for obesity (excluding liposuction > 1 year prior to enrolment)

Not pregnant/considering pregnancy

No functional impairments that prevents the performance of intensive exercise

Accept of medical regulation by the U-TURN endocrinologist

Inactivity, defined as < 1,5 hours of structured physical activity pr. week at moderate intensity and cycling < 30 minutes/5 km pr. day at moderate intensity (moderate intensity = out of breath but able to speak)

No participation in other research intervention studies

Exclusion Criteria:

HbA1c: >=75 mmol/mol with no glucose lowering medications

HbA1c: >=64 mmol/mol with mono glucose lowering therapy (if compliant with the prescription)

HbA1c: >=57 mmol/mol with >=dual glucose lowering therapy (if compliant with the prescription)

estimated glomerular filtration rate<60 mL/min

Protein or glucose in the urine at pre-screening

No biochemical sign of other major diseases

Presence of circulating glutamate-decarboxylase anti body (GAD) 65

Objective findings that contraindicates participation in intensive exercise

Anamnestic findings that contraindicates participation in the study

Unable to allocate the needed time to fulfill the intervention

Language barrier, mental incapacity, unwillingness or inability to understand and be able to complete the interventions

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: Single

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
No Intervention: Control
No intervention
Experimental: Dietary control (DCON)
The macro-nutrient distributions are in line with the current guidelines from the national Diabetes Association and Canadian guidelines, where individualization in macronutrient distribution should lie within the range of 45-60E% carbohydrate, 15-20E% protein and 20-35E% fat. The dietary plan will aim at reducing saturated fat intake <7E% aiming at a caloric deficit of 500 kilo calories/day
Dietary intervention
Experimental: Moderate Exercise Dose (MED)
Two aerobic training sessions per week of 45-60 min duration and one session per week with combined aerobic (30-35 min) and resistance (30 min) training and a dietary intervention (as above)
The participants will undergo diet or combined diet and exercise. The exercise will be provided at different volumes
Experimental: High Exercise Dose (HED)
Four aerobic training sessions per week of 45-60 min duration and two sessions per week with combined aerobic (30-35 min) and resistance (30 min) training and a dietary intervention (as above)
The participants will undergo diet or combined diet and exercise. The exercise will be provided at different volumes

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Pancreatic beta-cell function (Per protocol)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
The change in the late-phase disposition index (DI) during the final 30 minutes of hyperglycemic phase of the hyperglycemic clamp.
From baseline (0 weeks) to follow-up (16 weeks)

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Pancreatic beta-cell function (Intention to treat)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
As for per protocol
From baseline (0 weeks) to follow-up (16 weeks)
Glucagon like peptide 1 sensitivity (c-peptide)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Glucagon like peptide 1 stimulated C-peptide secretion
From baseline (0 weeks) to follow-up (16 weeks)
Glucagon like peptide 1 sensitivity (glucagon)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Glucagon like peptide 1 stimulated glucagon secretion
From baseline (0 weeks) to follow-up (16 weeks)
Glucagon like peptide 1 sensitivity (insulin)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Glucagon like peptide 1 stimulated insulin secretion
From baseline (0 weeks) to follow-up (16 weeks)
Arginine sensitivity (insulin)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Arginine stimulated insulin secretion
From baseline (0 weeks) to follow-up (16 weeks)
Arginine sensitivity (c-peptide)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Arginine stimulated C-peptide secretion
From baseline (0 weeks) to follow-up (16 weeks)
Arginine sensitivity (glucagon)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Arginine stimulated glucagon secretion
From baseline (0 weeks) to follow-up (16 weeks)
Early phase disposition index (c-peptide)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in 1st phase C-peptide secretion defined as the peak concentration during the initial 10 minutes of the hyperglycaemic clamp
From baseline (0 weeks) to follow-up (16 weeks)
Early phase disposition index (insulin)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in 1st phase insulin secretion defined as the peak concentration during the initial 10 minutes of the hyperglycaemic clamp
From baseline (0 weeks) to follow-up (16 weeks)
Glucose clearance
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Rate of glucose clearance (stable isotope infusion) during steady state hyperglycemia
From baseline (0 weeks) to follow-up (16 weeks)
Glucose appearance
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Rate of glucose appearance (stable isotope infusion) during steady state hyperglycemia
From baseline (0 weeks) to follow-up (16 weeks)
Insulin sensitivity
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in mean Glucose infusion rate over last 30 min of clamp phase/(mean insulin×glucose
From baseline (0 weeks) to follow-up (16 weeks)
Mean amplitude of glycemic excursions
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Mean amplitude of glycemic excursions (MAGE - calculated based on min 3 days sensor glucose profiles)
From baseline (0 weeks) to follow-up (16 weeks)
Coefficient of glucose variation
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Coefficient of variation defined as (mean glucose/the standard deviation (SD)) of min 3 days sensor glucose profiles
From baseline (0 weeks) to follow-up (16 weeks)
Mean glucose levels
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in the mean glucose levels (calculated based on min 3 days sensor glucose profiles)
From baseline (0 weeks) to follow-up (16 weeks)
Time in hyperglycemia
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in time in hyperglycaemia (calculated based on min 3 days sensor glucose profiles)
From baseline (0 weeks) to follow-up (16 weeks)
Time in hypoglycemia
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in time in hypoglycaemia from min 3 days sensor glucose profiles
From baseline (0 weeks) to follow-up (16 weeks)
Pancreatic fat
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Pancreatic fat
From baseline (0 weeks) to follow-up (16 weeks)
Hepatic fat
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Hepatic fat
From baseline (0 weeks) to follow-up (16 weeks)
Visceral fat
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in visceral fat
From baseline (0 weeks) to follow-up (16 weeks)
Total fat mass
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Total fat mass
From baseline (0 weeks) to follow-up (16 weeks)
Total fat free mass
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Total fat free mass
From baseline (0 weeks) to follow-up (16 weeks)
Total lean body mass
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Total lean body mass
From baseline (0 weeks) to follow-up (16 weeks)
Android fat mass
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in Android fat mass
From baseline (0 weeks) to follow-up (16 weeks)
Gynoid fat mass
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in gynoid fat mass
From baseline (0 weeks) to follow-up (16 weeks)
Body weight
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in body weight
From baseline (0 weeks) to follow-up (16 weeks)
Body mass index
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in body mass index
From baseline (0 weeks) to follow-up (16 weeks)
Systemic oxidative stress (RNA)
Time Frame: From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Change in 8-oxo-guanosine
From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Systemic oxidative stress (DNA)
Time Frame: From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Change in 8-oxo-deoxoguonase
From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Advanced glycation end-products (AGE)
Time Frame: From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Change in AGE
From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
The circulating receptor for advanced glycation end-products (sRAGE)
Time Frame: From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Change in sRAGE
From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Markers of low-grade inflammation
Time Frame: From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Change in inflammatory markers (e.g. high sensitive C-reactive protein, interferon-ϒ, interleukin-10, interleukin-8, interleukin-6, interleukin-1, TNFα)
From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Glycated haemoglobin type 1AC (HbA1c)
Time Frame: From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Change in HbA1c
From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Total cholesterol
Time Frame: From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Change in total cholesterol
From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Total triglyceride
Time Frame: From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Change in total triglyceride
From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Low density lipoprotein (LDL)
Time Frame: From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Change in LDL
From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
High density lipoprotein (HDL)
Time Frame: From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Change in HDL
From baseline (0 weeks) to follow-up (4, 12 and 16 weeks)
Systolic blood pressure
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change systolic blood pressures
From baseline (0 weeks) to follow-up (16 weeks)
Diastolic blood pressure
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change diastolic blood pressure
From baseline (0 weeks) to follow-up (16 weeks)
Glucose tolerance
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change incremental and total area under the curve (glucose, c-peptide, insulin) during a mixed meal tolerance test
From baseline (0 weeks) to follow-up (16 weeks)
Gastric emptying (AUC)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in the AUC (paracetamol) during a mixed meal tolerance test
From baseline (0 weeks) to follow-up (16 weeks)
Gastric emptying (Rate of appearance)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in rate of appearance of paracetamol during a mixed meal tolerance test
From baseline (0 weeks) to follow-up (16 weeks)
Physical fitness (VO2max)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in physical fitness (VO2) during a progressive maximal bicycle ergometer test
From baseline (0 weeks) to follow-up (16 weeks)
Muscular 1 repetition max (strength)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in 1 repetition max
From baseline (0 weeks) to follow-up (16 weeks)
Total physical activity
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in objectively measured physical activity (counts per minute)
From baseline (0 weeks) to follow-up (16 weeks)
Moderate and vigorous physical activity (MVPA)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in time spend on MVPA
From baseline (0 weeks) to follow-up (16 weeks)
Sedentary time (SED)
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in time spend on SED
From baseline (0 weeks) to follow-up (16 weeks)
Physical well being
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in physical well being (Based on the physical dimension score from short-form 36, range 0-100)
From baseline (0 weeks) to follow-up (16 weeks)
Mental well being
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in mental well being (Based on the mental dimension score from Short-form 36) (range 0-100)
From baseline (0 weeks) to follow-up (16 weeks)
Satiety
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in self-reported satiety (VAS) during a mixed meal tolerance test (range 0-10)
From baseline (0 weeks) to follow-up (16 weeks)

Other Outcome Measures

Outcome Measure
Measure Description
Time Frame
Muscular metabolic function
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in metabolic function (Based on muscle biopsies in a subset of participants, N=16-32))
From baseline (0 weeks) to follow-up (16 weeks)
Fat tissue metabolic function
Time Frame: From baseline (0 weeks) to follow-up (16 weeks)
Change in metabolic function (Based on muscle biopsies in a subset of participants, N=16-32)
From baseline (0 weeks) to follow-up (16 weeks)

Collaborators and Investigators

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

Investigators

  • Principal Investigator: Mathias Ried-Larsen, PhD, Centre for Physical Activity Research, Righospitalet

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)

December 12, 2018

Primary Completion (Anticipated)

August 1, 2021

Study Completion (Anticipated)

August 1, 2021

Study Registration Dates

First Submitted

November 22, 2018

First Submitted That Met QC Criteria

December 5, 2018

First Posted (Actual)

December 10, 2018

Study Record Updates

Last Update Posted (Actual)

December 13, 2018

Last Update Submitted That Met QC Criteria

December 12, 2018

Last Verified

December 1, 2018

More Information

Terms related to this study

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

Undecided

IPD Plan Description

If the data can be fully anonymized the data can be shared.

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

No

Studies a U.S. FDA-regulated device product

No

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 Diet

3
Subscribe