Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Maturitas ; 184: 107944, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38412750

ABSTRACT

Abnormal uterine bleeding is a frequent symptom in the perimenopause. Causes are numerous, ranging from physiological reactions due to decreasing/unstable ovarian function to premalignant and malignant conditions. Benign findings such as endometrial polyps and myomas increase with age, leading to more abnormal uterine bleeding in the perimenopause. Cervical and vaginal causes of abnormal uterine bleeding should be excluded by speculum examination. Sexually transmitted diseases or pregnancy should be ruled out. Measurement of haemoglobin and iron levels, human chorion gonadotropin and thyroid hormones are relevant in selected cases. Transvaginal ultrasound is an ideal first step for the evaluation of perimenopausal abnormal uterine bleeding. Saline or gel contrast sonohysterography improves the diagnostic accuracy. Based on the ultrasound findings, invasive procedures such as endometrial biopsy or hysteroscopy can be planned. Once premalignant and malignant causes are excluded, the necessity for treatment can be evaluated in collaboration with the patient. Heavy menstrual bleeding causing anaemia will need immediate treatment. In less severe cases and in intermenstrual bleeding, expectant management can be considered. Hormonal treatment, such as oral progestogens, combined oral contraceptives or insertion of the levonorgestrel intrauterine system, may be a possibility if anovulatory bleeding is interfering with quality of life. The amount of bleeding can be reduced both by antifibrinolytic and non-steroidal anti-inflammatory drugs, progestogens and the levonorgestrel intrauterine system. Focal intrauterine lesions such as endometrial polyps or submucous myomas may require operative hysteroscopic procedures. Endometrial ablation or endometrial resection are good choices in selected cases, but some women will need a hysterectomy to treat their abnormal uterine bleeding in perimenopause.


Subject(s)
Perimenopause , Uterine Hemorrhage , Humans , Female , Uterine Hemorrhage/etiology , Uterine Hemorrhage/therapy , Uterine Hemorrhage/diagnosis
3.
J Pediatr Rehabil Med ; 16(2): 369-380, 2023.
Article in English | MEDLINE | ID: mdl-36314228

ABSTRACT

PURPOSE: The Wii Balance Board (WBB) can be used for assessment of steady state balance (SSB), but its reliability has not been studied in children aged 6-9 years. This study aimed to determine the test-retest reliability of the WBB for measuring SSB in this population. A secondary aim was to determine the minimum detectable change (MDC) and standard error of measurement (SEM) of the WBB in children aged 6-9 years. METHODS: 52 children between 6-9 years of age participated. "One leg stand balance" was used to assess center of pressure velocity (COPV) and center of pressure area (COPA) on three occasions by the same tester. Two tests were conducted on the same day (Day 1) and the third test was performed on another day (Day 2), with a period of 5-13 days between the two test days. Intraclass correlation coefficient (ICC 3,1), SEMs, and MDC were calculated. RESULTS: Intra-day test-retest reliability of COPA was found to be good (ICC3,1 =0.86; 95% confidence interval [CI]: 0.75, 0.92) and that of COPV was also found to be good (ICC3,1 =0.87; 95% CI: 0.77, 0.92). Inter-day test-retest reliability was found to be good for COPA (ICC3,1 = 0.87; 95% CI: 0.75, 0.93) and COPV (ICC3,1 = 0.89; 95% CI: 0.81, 0.94). SEM for COPA in intra-day testing was 18.90 mm2 (15.78%), and in inter-day testing it was 16.44 mm2 (13.61%). SEM for COPV in intra-day testing was 1.12 mm/s (7.6%), and in inter-day testing it was 1.01 mm/s (6.9%). MDC for COPA in intra-day testing was 52.41mm2 (42.75%), and in inter-day testing was 45.58 mm2 (35.75%). MDC for COPV in intra-day testing was 3.11 mm/s (21.2%), and in inter-day testing it was 2.80 mm/s (18.9%). CONCLUSION: The WBB has good test-retest reliability for assessing SSB of children between 6-9 years. COPA measurements appear to be less sensitive to clinical changes in SSB when compared to COPV. Assessment of validity of the WBB in this age group is recommended before it can be considered as a potential balance assessment tool in children.


Subject(s)
Video Games , Humans , Child , Reproducibility of Results , Postural Balance
4.
Diabetes Obes Metab ; 24(2): 212-220, 2022 02.
Article in English | MEDLINE | ID: mdl-34595827

ABSTRACT

AIMS: To present secondary outcome analyses of liraglutide treatment in overweight adults with insulin pump-treated type 1 diabetes (T1D), focusing on changes in body composition and dimensions, and to evaluate changes in food intake to identify potential dietary drivers of liraglutide-associated weight loss. MATERIALS AND METHODS: A 26-week randomized placebo-controlled study was conducted to investigate the efficacy and safety of liraglutide 1.8 mg daily in 44 overweight adults with insulin pump-treated T1D and glucose levels above target, and demonstrated significant glycated haemoglobin (HbA1c)- and body weight-reducing effects. For secondary outcome analysis, dual X-ray absorptiometry scans were completed at Weeks 0 and 26, and questionnaire-based food frequency recordings were obtained at Weeks 0, 13 and 26 to characterize liraglutide-induced changes in body composition and food intake. RESULTS: Total fat and lean body mass decreased in liraglutide-treated participants (fat mass -4.6 kg [95% confidence interval {CI} -5.7; -3.5], P < 0.001; lean mass -2.5 kg [95% CI -3.2;-1.7], P < 0.001), but remained stable in placebo-treated participants (fat mass -0.3 kg [95% CI -1.3;0.8], P = 0.604; lean mass 0.0 kg [95% CI -0.7;0.7]; P = 0.965 [between-group P values <0.001]). Participants reduced their energy intake numerically more in the liraglutide arm (-1.1 MJ [95% CI -2.0;-0.02], P = 0.02) than in the placebo arm (-0.9 MJ [95% CI -2.0;0.1], P = 0.22), but the between-group difference was statistically insignificant (P = 0.42). However, energy derived from added sugars decreased by 27% in the liraglutide arm compared with an increase of 14% in the placebo arm (P = 0.004). CONCLUSIONS: Liraglutide lowered fat and lean body mass compared with placebo. Further, liraglutide reduced intake of added sugars. However, no significant difference in total daily energy intake was detected between liraglutide- and placebo-treated participants.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Insulins , Adult , Body Composition , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Double-Blind Method , Drug Therapy, Combination , Glycated Hemoglobin/metabolism , Humans , Hypoglycemic Agents/adverse effects , Liraglutide/adverse effects , Overweight/complications , Overweight/drug therapy , Sugars/therapeutic use , Treatment Outcome
5.
Entropy (Basel) ; 22(1)2020 Jan 09.
Article in English | MEDLINE | ID: mdl-33285854

ABSTRACT

Previous literature has demonstrated that hypoglycemic events in patients with type 1 diabetes (T1D) are associated with measurable scalp electroencephalography (EEG) changes in power spectral density. In the present study, we used a dataset of 19-channel scalp EEG recordings in 34 patients with T1D who underwent a hyperinsulinemic-hypoglycemic clamp study. We found that hypoglycemic events are also characterized by EEG complexity changes that are quantifiable at the single-channel level through empirical conditional and permutation entropy and fractal dimension indices, i.e., the Higuchi index, residuals, and tortuosity. Moreover, we demonstrated that the EEG complexity indices computed in parallel in more than one channel can be used as the input for a neural network aimed at identifying hypoglycemia and euglycemia. The accuracy was about 90%, suggesting that nonlinear indices applied to EEG signals might be useful in revealing hypoglycemic events from EEG recordings in patients with T1D.

6.
Lancet Diabetes Endocrinol ; 8(4): 313-324, 2020 04.
Article in English | MEDLINE | ID: mdl-32135138

ABSTRACT

BACKGROUND: In type 2 diabetes, long-acting GLP-1 receptor agonists lower fasting plasma glucose and improve glycaemic control via their insulinotropic and glucagonostatic effects. In type 1 diabetes, their efficacy as an add-on treatment to insulin therapy is modest. Short-acting GLP-1 receptor agonists also lower postprandial glucose excursions in type 2 diabetes by decelerating gastric emptying rate. We aimed to test the efficacy of a short-acting GLP-1 receptor agonist in type 1 diabetes. METHODS: In the single-centre, parallel-group, randomised, double-blind, placebo-controlled MAG1C trial, patients with type 1 diabetes on multiple daily injection therapy aged 18 years and older with HbA1c 59-88 mmol/mol (7·5-10·0%) and a BMI of more than 22·0 kg/m2 were randomly assigned (1:1) through a computer-generated randomisation list to preprandial subcutaneous injection of 10 µg exenatide (Byetta) or placebo three times daily for 26 weeks as an add-on treatment to usual insulin therapy. Clinically assessed insulin titration was done by study staff. Participants and investigators were masked to treatment allocation. The primary endpoint was between-group difference in HbA1c after 26 weeks. Data were analysed with a baseline-adjusted linear mixed model in the intention-to-treat population. This study is registered with ClinicalTrials.gov, NCT03017352, and is completed. FINDINGS: Between Jan 4, 2017, and Jan 16, 2019, 108 participants were randomly assigned, 54 to exenatide and 54 to placebo; 23 participants discontinued treatment (17 in the exenatide group and six in the placebo group). From a baseline-adjusted mean of 66·4 mmol/mol (95% CI 64·9-67·8 [8·2%, 8·1-8·4]), HbA1c changed by -3·2 mmol/mol (-5·0 to -1·4 [-0·3%, -0·5 to -0·1]) with exenatide and -2·1 mmol/mol (-3·7 to -0·6 [-0·2%, -0·3 to -0·1]) with placebo after 26 weeks (estimated treatment difference of -1·1 mmol/mol (-3·4 to 1·2 [-0·1%, -0·3 to 0·1]; p=0·36). Exenatide increased the number of self-reported gastrointestinal adverse events (primarily nausea [48 events among 37 patients with exenatide, nine with placebo among 9 patients]). Two serious adverse events occurred in the exenatide group, and six occurred in the placebo group (none were considered to be related to the study drug). INTERPRETATION: Short-acting exenatide does not seem to have a future as a standard add-on treatment to insulin therapy in type 1 diabetes. FUNDING: AstraZeneca.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Type 1/blood , Exenatide/therapeutic use , Glycated Hemoglobin/drug effects , Hypoglycemic Agents/therapeutic use , Meals/physiology , Adult , Body Mass Index , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/physiopathology , Double-Blind Method , Female , Glucagon-Like Peptides , Humans , Male , Treatment Outcome
7.
Diabetes Obes Metab ; 22(4): 492-500, 2020 04.
Article in English | MEDLINE | ID: mdl-31696598

ABSTRACT

AIM: To investigate the efficacy of adding the glucagon-like peptide-1 receptor agonist liraglutide to continuous subcutaneous insulin infusion (CSII) in overweight or obese persons with type 1 diabetes and non-optimal glycaemic control. MATERIALS AND METHODS: A 26-week, randomized, double-blind, placebo-controlled trial including 44 overweight or obese adults with type 1 diabetes randomized 1:1 to liraglutide 1.8 mg once daily (QD) or placebo added to CSII treatment. The primary endpoint was change in haemoglobin A1c (HbA1c). Secondary endpoints included change in insulin dose, CSII settings, glycaemic variability, body weight and patient-reported outcome measures. Finally, adverse effects including hypoglycaemic events were registered. RESULTS: HbA1c was reduced by 5 mmol/mol (0.5%) from a baseline of 66 mmol/mol (8.2%) in patients treated with liraglutide compared with a non-significant change of +2.3 mmol/mol (0.2%) from a baseline of 66 mmol/mol (8.1%) in patients treated with placebo (between-group difference 7 mmol/mol [0.7%], P < 0.001). Liraglutide reduced total insulin dose by 8 units/day or 16% of total insulin dose (P = 0.008). Mean body weight was reduced by 6.3 kg (P < 0.001) compared with placebo. Concomitantly, time spent in glycaemic target range 4-10 mmol/L (71-180 mg/dL) increased while the risk of hypoglycaemia did not differ between groups at the end of treatment. CONCLUSION: Liraglutide treatment reduced HbA1c, total daily insulin dose and body weight without increasing the risk of hypoglycaemia in CSII-treated patients with type 1 diabetes and insufficient glycaemic control. Liraglutide may be considered a potential add-on therapy to insulin in this subgroup of patients.


Subject(s)
Diabetes Mellitus, Type 1 , Hyperglycemia , Adult , Body Weight , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Double-Blind Method , Drug Therapy, Combination , Glycated Hemoglobin , Humans , Hyperglycemia/prevention & control , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Liraglutide/therapeutic use , Overweight/complications , Treatment Outcome
8.
Article in English | MEDLINE | ID: mdl-31031712

ABSTRACT

The role of the autonomic nervous system in the efficacy of glucagon-like peptide-1 receptor agonists (GLP-1 RA) in patients with type 1 diabetes is unknown. We assessed the association between autonomic function and weight loss induced by the GLP-1 RA liraglutide. Methods: Lira-1 was a randomized, double-blind, placebo-controlled trial assessing the efficacy and safety of 1.8 mg liraglutide once-daily for 24 weeks in overweight patients with type 1 diabetes. Autonomic function was assessed by heart rate response to deep breathing (E/I ratio), to standing (30/15 ratio), to the Valsalva maneuver and resting heart rate variability (HRV) indices. Associations between baseline the cardiovascular autonomic neuropathy (CAN) diagnosis (> 1 pathological non-resting test) and levels of test outcomes on liraglutide-induced weight loss was assessed by linear regression models. Results: Ninety-nine patients with mean age 48 (SD 12) years, HbA1c 70 (IQR 66;75) mmol/mol and BMI of 30 (SD 3) kg/m2 were assigned to liraglutide (N = 50) or placebo (N = 49). The CAN diagnosis was not associated with weight loss. A 50% higher baseline level of the 30/15 ratio was associated with a larger weight reduction by liraglutide of -2.65 kg during the trial (95% CI: -4.60; -0.69; P = 0.009). Similar significant associations were found for several HRV indices. Conclusions: The overall CAN diagnosis was not associated with liraglutide-induced weight loss in overweight patients with type 1 diabetes. Assessed separately, better outcomes for several CAN measures were associated with higher weight loss, indicating that autonomic involvement in liraglutide-induced weight loss may exist.

9.
Int J Cardiol ; 278: 280-284, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30291010

ABSTRACT

BACKGROUND: Long duration of diabetes mellitus (DM) is associated with an increased risk of infection, however no studies have yet focused on the duration of DM and the associated risk of infective endocarditis (IE). METHODS: Patients with DM were identified through the Danish Prescription Registry, 1996-2015. Duration of DM was split in follow-up periods of: 0-5 years, 5-10 years, 10-15 years, and >15 years. Multivariable adjusted Poisson regression was used to calculate incidence rate ratios (IRR) according to study groups. DM late-stage complications and the associated risk of IE were investigated as time-varying covariates using the validated Diabetes Complications Severity Index (DCSI). RESULTS: We included 299,551 patients with DM. In patients with DM duration of 0-5 years, 5-10 years, 10-15 years, and >15 years, the incidence rates of IE were 0.24, 0.33, 0.58, and 0.96 cases of IE/1000 person years, respectively. Patients with DM duration 5-10 years, 10-15 years, and >15 years were associated with a higher risk of IE with an IRR of 1.24 (95% CI: 1.02-1.51), 1.92 (95% CI: 1.52-2.43) and 3.05 (95% CI: 2.11-4.40), respectively, compared with DM duration 0-5 years. Patients with a DCSI score of 2, 3 and >3 were associated with a higher risk of IE compared with patients with a DCSI score of 0, IRR = 1.78 (95% CI: 1.34-2.36), IRR = 2.34 (95% CI: 1.73-3.16), and IRR = 2.59 (95% CI: 1.92-3.48), respectively. CONCLUSION: This study shows a stepwise increase in the risk of IE with DM duration and severity independent of age and known comorbidity.


Subject(s)
Diabetes Complications/diagnosis , Diabetes Complications/epidemiology , Diabetes Mellitus/diagnosis , Diabetes Mellitus/epidemiology , Endocarditis/diagnosis , Endocarditis/epidemiology , Aged , Denmark/epidemiology , Diabetes Complications/blood , Diabetes Mellitus/blood , Endocarditis/blood , Female , Follow-Up Studies , Hospitalization/trends , Humans , Male , Middle Aged , Registries , Risk Factors , Time Factors
10.
Expert Opin Pharmacother ; 19(9): 947-960, 2018 06.
Article in English | MEDLINE | ID: mdl-29991320

ABSTRACT

INTRODUCTION: Despite intensified insulin treatment, many persons with type 1 diabetes (T1D) do not achieve glycemic and metabolic targets. Consequently, non-insulin chemical therapies that improve glycemic control and metabolic parameters without increasing the risk of adverse events (including hypoglycemia) are of interest as adjunct therapies to insulin. AREAS COVERED: In this review, the authors discuss the efficacy and safety of non-insulin therapies, including pramlintide, glucagon-like peptide-1 (GLP-1) receptor agonists, dipeptidyl peptidase-4 inhibitors (DPP-4), sodium-glucose cotransporter (SGLT1 and SGLT2) inhibitors, metformin, sulfonylureas, and thiazolidinediones as add-on therapies to insulin in T1D. EXPERT OPINION: The current evidence shows that the efficacy of non-insulin therapies as add-on therapies to insulin is minimal or modest with an average HbA1c reduction of 0.2-0.5% (2-6 mmol/mol). Indeed, the current focus is on the development of SGLT inhibitors as adjuncts to insulin in type 1 diabetes. Studies of subgroups with obesity, residual beta-cell function (including newly diagnosed patients) and patients prone to hypoglycemia could be areas of future research.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/therapeutic use , Clinical Trials as Topic , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Gastrointestinal Diseases/etiology , Humans , Hypoglycemic Agents/adverse effects , Islet Amyloid Polypeptide/therapeutic use , Metformin/therapeutic use , Sulfonylurea Compounds/therapeutic use , Thiazolidinediones/therapeutic use
11.
Qual Life Res ; 27(1): 267-271, 2018 01.
Article in English | MEDLINE | ID: mdl-28677076

ABSTRACT

PURPOSE: The aim of this study was to translate and culturally adapt the Short Musculoskeletal Function Assessment (SMFA) into Danish (SMFA-DK) and assess the psychometric properties. METHODS: SMFA was translated and cross-culturally adapted according to a standardized procedure. Minor changes in the wording in three items were made to adapt to Danish conditions. Acute patients (n = 201) and rehabilitation patients (n = 231) with musculoskeletal problems aged 18-87 years were included. The following analysis were made to evaluate psychometric quality of SMFA-DK: Reliability with Chronbach's alpha, content validity as coding according to the International Classification of Functioning, Disability and Health (ICF), floor/ceiling effects, construct validity as factor analysis, correlations between SMFA-DK and Short Form 36 and also known group method. Responsiveness and effect size were calculated. RESULTS: Cronbach's alpha values were between 0.79 and 0.94. SMFA-DK captured all components of the ICF, and there were no floor/ceiling effects. Factor analysis demonstrated four subscales. SMFA-DK correlated good with the SF-36 subscales for the rehabilitation patients and lower for the newly injured patients. Effect sizes were excellent and better for SMFA-DK than for SF-36. CONCLUSION: The study indicates that SMFA-DK can be a valid and responsive measure of outcome in rehabilitation settings.


Subject(s)
Cross-Cultural Comparison , Disability Evaluation , Musculoskeletal Diseases/diagnosis , Psychometrics/methods , Quality of Life/psychology , Aged , Female , Humans , Male , Middle Aged , Musculoskeletal Diseases/pathology , Reproducibility of Results , Surveys and Questionnaires , Sweden
12.
Expert Rev Clin Pharmacol ; 10(6): 621-632, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28349716

ABSTRACT

INTRODUCTION: A fixed combination of basal insulin degludec and glucagon-like peptide-1 receptor agonist (GLP-1RA) liraglutide (IDegLira; 50 units degludec/1.8 mg liraglutide) has been developed as a once daily injection for the treatment of type 2 diabetes (T2D). In the phase 3a trial programme 'Dual action of liraglutide and insulin degludec in type 2 diabetes' (DUAL™), five trials of 26 weeks duration and one trial of 32 weeks duration have evaluated the efficacy and safety of IDegLira compared with administration of insulin degludec, insulin glargine, liraglutide alone or placebo. Areas covered: Combination therapy with IDegLira reduces HbA1c more than monotherapy with a GLP-1RA (liraglutide) or insulin (degludec or glargine). Combination therapy leads also to weight loss, or a stable body weight, with no increase in hypoglycaemia. Rates of adverse events did not differ between treatment groups; however, gastrointestinal side effects were fewer with IDegLira compared with liraglutide treatment alone. A limitation of the DUAL™ development programme is that patients receiving basal insulin doses in excess of 50 units were excluded from the studies. Expert commentary: In conclusion, IDegLira combines the clinical advantages of basal insulin and GLP-1RA treatment, and is a treatment strategy that could improve the management of patients with T2D.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin, Long-Acting/therapeutic use , Liraglutide/therapeutic use , Drug Combinations , Glucagon-Like Peptide-1 Receptor/agonists , Glycated Hemoglobin/metabolism , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacology , Insulin, Long-Acting/adverse effects , Insulin, Long-Acting/pharmacology , Liraglutide/adverse effects , Liraglutide/pharmacology , Weight Loss/drug effects
13.
Diabetes Technol Ther ; 19(2): 85-90, 2017 02.
Article in English | MEDLINE | ID: mdl-28118048

ABSTRACT

BACKGROUND: Recurrent hypoglycemia has been shown to blunt hypoglycemia symptom scores and counterregulatory hormonal responses during subsequent hypoglycemia. We therefore studied whether hypoglycemia-associated electroencephalogram (EEG) changes are affected by an antecedent episode of hypoglycemia. METHODS: Twenty-four patients with type 1 diabetes mellitus (10 with normal hypoglycemia awareness, 14 with hypoglycemia unawareness) were studied on 2 consecutive days by hyperinsulinemic glucose clamp at hypoglycemia (2.0-2.5 mmol/L) during a 1-h period. EEG was recorded, cognitive function assessed, and hypoglycemia symptom scores and counterregulatory hormonal responses were obtained. RESULTS: Twenty-one patients completed the study. Hypoglycemia-associated EEG changes were identified on both days with no differences in power or frequency distribution in the theta, alpha, or the combined theta-alpha band during hypoglycemia on the 2 days. Similar degree of cognitive dysfunction was also present during hypoglycemia on both days. When comparing the aware and unaware group, there were no differences in the hypoglycemia-associated EEG changes. There were very subtle differences in cognitive function between the two groups on day 2. The symptom response was higher in the aware group on both days, while only subtle differences were seen in the counterregulatory hormonal response. CONCLUSION: Antecedent hypoglycemia does not affect hypoglycemia-associated EEG changes in patients with type 1 diabetes mellitus.


Subject(s)
Blood Glucose/analysis , Brain/physiopathology , Cognition/physiology , Diabetes Mellitus, Type 1/physiopathology , Hypoglycemia/physiopathology , Adult , Diabetes Mellitus, Type 1/drug therapy , Electroencephalography , Female , Glucose Clamp Technique , Humans , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Male , Middle Aged
14.
Diabetes Obes Metab ; 19(5): 734-738, 2017 05.
Article in English | MEDLINE | ID: mdl-27935240

ABSTRACT

We investigated the short-term effect of adding liraglutide 1.8 mg once daily to insulin treatment on cardiovascular risk factors in patients with type 1 diabetes. In total, 100 overweight (BMI ≥25 kg/m2 ) adult patients (age ≥18 years) with type 1 diabetes and HbA1c ≥ 8% (64 mmol/mol) were randomized to liraglutide 1.8 mg or placebo added to insulin treatment in a 24-week double-blinded, placebo-controlled trial. At baseline and after 24 weeks of treatment, 24-hour blood pressure and heart rate, pulse pressure, pulse wave velocity and carotid intima-media thickness were evaluated. Compared with placebo, liraglutide increased 24-hour heart rate by 4.6 beats per minute (BPM); P = .0015, daytime heart rate by 3.7; P = .0240 and night-time heart rate by 7.5 BPM; P < .001 after 24 weeks. Diastolic nocturnal blood pressure increased by 4 mm Hg; P = .0362 in the liraglutide group compared with placebo. In conclusion, in patients with long-standing type 1 diabetes, liraglutide as add-on to insulin increased heart rate and did not improve other cardiovascular risk factors after 24 weeks of treatment.


Subject(s)
Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Type 1/drug therapy , Diabetic Angiopathies/prevention & control , Diabetic Cardiomyopathies/prevention & control , Hypoglycemic Agents/adverse effects , Liraglutide/adverse effects , Adult , Blood Pressure/drug effects , Body Mass Index , Cardiovascular Diseases/complications , Cardiovascular Diseases/diagnostic imaging , Cardiovascular Diseases/epidemiology , Carotid Intima-Media Thickness , Denmark/epidemiology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/complications , Diabetic Angiopathies/diagnostic imaging , Diabetic Angiopathies/epidemiology , Diabetic Cardiomyopathies/diagnostic imaging , Diabetic Cardiomyopathies/epidemiology , Double-Blind Method , Drug Resistance , Drug Therapy, Combination/adverse effects , Female , Glycated Hemoglobin/analysis , Heart Rate/drug effects , Humans , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Liraglutide/therapeutic use , Male , Middle Aged , Overweight/complications , Pulse Wave Analysis , Risk Factors
15.
Diabetes Obes Metab ; 19(6): 773-782, 2017 06.
Article in English | MEDLINE | ID: mdl-27868372

ABSTRACT

AIM: Glucagon-like peptide-1 receptor agonist (GLP-1RA) therapy is a potential treatment as adjunct to insulin in type 1 diabetes (T1D). However, GLP-1RAs inhibit glucagon secretion and delay the gastric emptying (GE) rate and may impair recovery from hypoglycaemia. We evaluated the effect of the GLP-1RA liraglutide on counterregulatory responses and GE rate during hypoglycaemia in persons with T1D. MATERIALS AND METHODS: In a 12-week, randomized, double-blind, placebo-controlled study, 20 patients aged >18 years with T1D and HbA1c ≥8% (64 mmol/mol) were randomly assigned (1:1) to liraglutide 1.2 mg once daily or placebo as add-on to insulin treatment. Before and at end of treatment a hypoglycaemic clamp (plasma glucose target 2.5 mmol/L) was carried out, followed by a liquid meal. Primary endpoint was change in GE rate (evaluated by area under the paracetamol curve and time to peak). Secondary endpoints included changes in glycaemic recovery, counter-regulatory hormones, pancreatic polypeptide (PP), GLP-1, blood pressure and heart rate. RESULTS: During the period June 2013 to October 2014, 20 patients were enrolled. After 12 weeks of treatment, changes in GE rates did not differ significantly between groups ( P = .96), with no significant changes from baseline, whether evaluated from AUCs or time to peak. The secondary endpoints, glycaemic recovery, counter-regulatory hormone responses, systolic blood pressure and GLP-1 and PP responses, were also similar. Heart rate increased with liraglutide from 69 ± 4 to 80 ± 5 beats/min ( P = .02). CONCLUSIONS: Liraglutide does not compromise glycaemic recovery, GE rate or counter-regulatory hormone responses in T1D patients during hypoglycaemia. No treatment-related safety issues were identified.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Hypoglycemia/drug therapy , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Liraglutide/administration & dosage , Adult , Aged , Blood Glucose/drug effects , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/complications , Double-Blind Method , Drug Therapy, Combination , Female , Gastric Emptying/drug effects , Glycated Hemoglobin/drug effects , Humans , Hypoglycemia/etiology , Hypoglycemia/physiopathology , Hypoglycemic Agents/adverse effects , Insulin/adverse effects , Liraglutide/adverse effects , Male , Middle Aged , Research Design , Treatment Outcome , Young Adult
16.
Lancet Diabetes Endocrinol ; 4(9): 766-780, 2016 09.
Article in English | MEDLINE | ID: mdl-26969516

ABSTRACT

Insulin treatment of individuals with type 1 diabetes has shortcomings and many patients do not achieve glycaemic and metabolic targets. Consequently, the focus is on novel non-insulin therapeutic approaches that reduce hyperglycaemia and improve metabolic variables without increasing the risk of hypoglycaemia or other adverse events. Several therapies given in conjunction with insulin have been investigated in clinical trials, including pramlintide, glucagon-like peptide-1 receptor agonists, dipeptidyl peptidase-4 inhibitors, sodium-glucose co-transporter inhibitors, metformin, sulfonylureas, and thiazolidinediones. These drugs have pleiotropic effects on glucose metabolism and different actions complementary to those of insulin-this Review reports the effects of these drugs on glycaemic control, glucose variability, hypoglycaemia, insulin requirements, and bodyweight. Existing studies are of short duration with few participants; evidence for the efficacy of concomitant treatments is scarce and largely clinically insignificant. A subgroup of patients with type 1 diabetes for whom non-insulin antidiabetic drugs could significantly benefit glycaemic control cannot yet be defined, but we suggest that obese patients prone to hypoglycaemia and patients with residual ß-cell function are populations of interest for future trials.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Hyperglycemia/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Diabetes Mellitus, Type 1/complications , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Glucagon-Like Peptide-1 Receptor/agonists , Humans , Hyperglycemia/etiology , Islet Amyloid Polypeptide/therapeutic use , Metformin/therapeutic use , Sodium-Glucose Transporter 2 , Sodium-Glucose Transporter 2 Inhibitors , Sulfonylurea Compounds/therapeutic use , Thiazolidinediones/therapeutic use
17.
Expert Opin Biol Ther ; 16(4): 579-90, 2016.
Article in English | MEDLINE | ID: mdl-26926662

ABSTRACT

INTRODUCTION: Many persons with type 1 diabetes do not achieve glycemic targets, why new treatments, complementary to insulin, are of interest. Liraglutide, a long-acting glucagon-like peptide-1 receptor agonist could be a potential pharmacological supplement to insulin. This review discusses the mechanism of actions, efficacy and safety of liraglutide as add-on to insulin in persons with type 1 diabetes. AREAS COVERED: Physiological and clinical data on liraglutide in type 1 diabetes were reviewed. We searched the Cochrane library, MEDLINE and EMBASE, with the final search performed February 16, 2016. EXPERT OPINION: Liraglutide as adjunct to insulin treatment reduced body weight and daily dose of insulin compared with insulin alone. The effect on HbA1c was inconsistent with mostly uncontrolled, small-scale studies reporting improvements in glycemic control. In placebo-controlled studies there was no clinically relevant effect on HbA1c. Adverse events were mostly transient gastrointestinal side effects, primarily nausea. Based on the available data, liraglutide cannot be recommended as add-on therapy to insulin in persons with type 1 diabetes with the aim to improve glycemic control. Ongoing trials in newly diagnosed patients with type 1 diabetes and in insulin pump-treated patients will help define the future role of liraglutide therapy in type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/therapeutic use , Incretins/therapeutic use , Liraglutide/therapeutic use , Blood Glucose/drug effects , Body Weight , Glucagon-Like Peptide-1 Receptor/agonists , Humans
18.
Expert Rev Clin Pharmacol ; 9(2): 241-65, 2016.
Article in English | MEDLINE | ID: mdl-26573176

ABSTRACT

Over the last decade, the discovery of glucagon-like peptide 1 receptor agonists (GLP-1 RAs) has increased the treatment options for patients with type 2 diabetes mellitus (T2DM). GLP-1 RAs mimic the effects of native GLP-1, which increases insulin secretion, inhibits glucagon secretion, increases satiety and slows gastric emptying. This review evaluates the phase III trials for all approved GLP-1 RAs and reports that all GLP-1 RAs decrease HbA1c, fasting plasma glucose, and lead to a reduction in body weight in the majority of trials. The most common adverse events are nausea and other gastrointestinal discomfort, while hypoglycaemia is rarely reported when GLP-1 RAs not are combined with sulfonylurea or insulin. Treatment options in the near future will include co-formulations of basal insulin and a GLP-1 RA.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/therapeutic use , Animals , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/physiopathology , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin Secretion
19.
Lancet Diabetes Endocrinol ; 4(3): 221-232, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26656289

ABSTRACT

BACKGROUND: The combination of insulin and glucagon-like peptide-1 (GLP-1) receptor agonist therapy improves glycaemic control, induces weight loss, and reduces insulin dose needed in type 2 diabetes. We assessed the efficacy and safety of the GLP-1 receptor agonist liraglutide as an add-on therapy to insulin for overweight adult patients with type 1 diabetes. METHODS: We did a randomised, double-blind, placebo-controlled trial at Steno Diabetes Center (Gentofte, Denmark). Patients aged 18 years or older with type 1 diabetes, insufficient glycaemic control (HbA1c >8% [64 mmol/mol]), and overweight (BMI >25 kg/m(2)) were randomly assigned (1:1) to receive insulin treatment plus either liraglutide or placebo (saline solution) by subcutaneous injection once per day. Randomisation was done in blocks of four. Treatment assignment was masked to investigators and patients. Treatment lasted 24 weeks and liraglutide was started at a dose of 0·6 mg per day, escalated to 1·2 mg per day after 1 week, and then again to 1·8 mg per day after another week. Intervals between dose increments could be extended at the discretion of the investigator. The primary endpoint was change in HbA1c from baseline to week 24. Secondary endpoints were changes in hypoglycaemic events, glycaemic variability, glycaemic excursions, insulin dose, bodyweight, postprandial plasma concentrations of glucagon and GLP-1, gastric emptying, blood pressure, heart rate, patient-reported outcome measures, time spent in hypoglycaemia, near-normoglycaemia, and hyperglycaemia, plasma fasting glucose, mean glucose, and cholesterol. Efficacy analyses were calculated by use of a mixed model, whereby a patient's data are used as long as the patient is in the study. The safety analyses were done in the intention-to-treat population, which consisted of all patients who received at least one dose of their randomly assigned study drug. This study is registered with ClinicalTrials.gov, number NCT01612468. FINDINGS: Between July 10, 2012, and May 30, 2014, we enrolled 100 patients with type 1 diabetes, with 50 patients allocated liraglutide and 50 to placebo. Four patients from the liraglutide group and six patients from the placebo group discontinued treatment before 24 weeks. At the end of treatment, change in HbA1c from baseline did not differ between groups (-0·5%, 95% CI -0·8 to -0·4 [-6·0 mmol/mol, 95% CI -8·7 to -4·4] with liraglutide vs -0·3%, -0·6 to -0·2 [-4·0 mmol/mol, -6·6 to -2·3] with placebo; between-group difference -0·2% [-0·5 to 0·1; 2·2 mmol/mol, -5·5 to 1·1], p=0·1833). The number of hypoglycaemic events was reduced with liraglutide, with an incident rate ratio of 0·82 (95% CI 0·74 to 0·90). However, we detected no changes in glycaemic variability (continuous overall net glycaemic action per 60 min from 10·3 [95% CI 9·8 to 10·8] to 9·9 [9·2 to 10·6] in the liraglutide treated patients vs 10·2 [9·7 to 10·7] to 9·7 [9·1 to 10·3] in the placebo treated patients). Both bolus insulin (difference -5·8 IU, 95% CI -10·7 to -0·8, p=0·0227) and bodyweight (difference -6·8 kg, 95% CI -12·2 to -1·4, p=0·0145) decreased with liraglutide treatment compared with placebo. Heart rate increased with liraglutide, with a difference between groups of 7·5 bpm (95% CI 2·8-12·2, p=0·0019). Postprandial plasma glucagon and GLP-1 concentrations did not differ between groups (difference between groups at end of treatment: -408 mmol/L per 240 min [95% CI -941 to 125, p=0·1309] for glucagon and -266 mmol/L per 240 min [-1034 to 501, p=0·4899] for GLP-1). Gastric emptying was delayed after 3 weeks of treatment with liraglutide (19·9 min, 95% CI 0·8 to 39·0, p=0·0412), but we detected no difference after 24 weeks of treatment (-1·5 min, -20·5 to 17·6, p=0·8793). Patient-reported outcome measures differed between groups only with respect to perceived frequency of hypoglycaemia, which was higher with placebo, with a difference between groups of -0·6 (95% CI -1·1 to -0·07, p=0·0257). Liraglutide was associated with more frequent nausea (29 [58%] patients with liraglutide vs five [10%] with placebo), dyspepsia (11 [22%] patients with liraglutide vs one [2%] with placebo), diarrhoea (ten [20%] patients with liraglutide vs one [2%] with placebo), decreased appetite (seven patients [14%] with liraglutide vs none with placebo), and vomiting (seven [14%] patients with liraglutide vs one [2%] with placebo). INTERPRETATION: In patients with type 1 diabetes, overweight, and insufficient glycaemic control, the reduction in HbA1c did not differ between insulin plus placebo and insulin plus liraglutide treatment. Liraglutide was associated with reductions in hypoglycaemic events, bolus and total insulin dose, and bodyweight, and increased heart rate. FUNDING: Novo Nordisk.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Liraglutide/therapeutic use , Overweight/complications , Blood Pressure/drug effects , Body Weight/drug effects , Double-Blind Method , Drug Therapy, Combination , Female , Glycated Hemoglobin/metabolism , Humans , Hypoglycemic Agents/adverse effects , Liraglutide/adverse effects , Male , Middle Aged , Treatment Outcome
20.
Diabetes Care ; 38(12): 2250-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26486191

ABSTRACT

OBJECTIVE: This study investigated the efficacy and safety of once-daily liraglutide 1.2 mg versus placebo as add-on to insulin treatment in normal-weight patients with poorly controlled type 1 diabetes. RESEARCH DESIGN AND METHODS: In a randomized (1:1), double-blind, placebo-controlled design, 40 patients with type 1 diabetes (HbA1c ≥8% [64 mmol/mol]) received once-daily liraglutide 1.2 mg or placebo for 12 weeks. Continuous glucose monitoring was performed before and at the end of treatment. The primary end point was change in HbA1c. Secondary end points included change in insulin dose, weight, glycemic excursions, heart rate, and blood pressure. RESULTS: Baseline HbA1c was similar in the liraglutide and placebo group (8.8 ± 0.2 and 8.7 ± 0.1% [72.5 ± 2.2 and 71.8 ± 1.5 mmol/mol]). Change in HbA1c from baseline was -0.6 ± 0.2% (-6.22 ± 1.71 mmol/mol) with liraglutide and -0.5 ± 0.2% (-5.56 ± 1.67 mmol/mol) with placebo (P = 0.62). Variation in glycemic excursions did not change in either group. Change in body weight was -3.13 ± 0.58 and +1.12 ± 0.42 kg (P < 0.0001) with liraglutide and placebo, respectively. The bolus insulin dose decreased in liraglutide-treated patients and did not change with placebo treatment (4.0 ± 1.3 vs. 0.0 ± 1.0 IU, P = 0.02). Heart rate increased within the liraglutide group (P = 0.04) but not compared with placebo, whereas mean systolic blood pressure decreased compared with placebo (between-group difference 3.21 mmHg [95% CI -8.31 to 1.90], P = 0.04). Liraglutide was more frequently associated with gastrointestinal adverse effects. The incidence of hypoglycemia did not differ between groups. CONCLUSIONS: Liraglutide significantly reduces body weight and insulin requirements but has no additional effect on HbA1c in normal-weight patients with type 1 diabetes inadequately controlled on insulin alone.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Liraglutide/therapeutic use , Adult , Body Weight , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Hypoglycemic Agents/administration & dosage , Liraglutide/administration & dosage , Male , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...