Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
JMIR Hum Factors ; 11: e55571, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38888941

RESUMO

BACKGROUND: The high number of unnecessary alarms in intensive care settings leads to alarm fatigue among staff and threatens patient safety. To develop and implement effective and sustainable solutions for alarm management in intensive care units (ICUs), an understanding of staff interactions with the patient monitoring system and alarm management practices is essential. OBJECTIVE: This study investigated the interaction of nurses and physicians with the patient monitoring system, their perceptions of alarm management, and smart alarm management solutions. METHODS: This explorative qualitative study with an ethnographic, multimethods approach was conducted in an ICU of a German university hospital. Using triangulation in data collection, 102 hours of field observations, 12 semistructured interviews with ICU staff members, and the results of a participatory task were analyzed. The data analysis followed an inductive, grounded theory approach. RESULTS: Nurses and physicians reported interacting with the continuous vital sign monitoring system for most of their work time and tasks. There were no established standards for alarm management; instead, nurses and physicians stated that alarms were addressed through ad hoc reactions, a practice they viewed as problematic. Staff members' perceptions of intelligent alarm management varied, but they highlighted the importance of understandable and traceable suggestions to increase trust and cognitive ease. CONCLUSIONS: Staff members' interactions with the omnipresent patient monitoring system and its alarms are essential parts of ICU workflows and clinical decision-making. Alarm management standards and workflows have been shown to be deficient. Our observations, as well as staff feedback, suggest that changes are warranted. Solutions for alarm management should be designed and implemented with users, workflows, and real-world data at the core.


Assuntos
Alarmes Clínicos , Unidades de Terapia Intensiva , Pesquisa Qualitativa , Humanos , Alemanha , Masculino , Feminino , Adulto , Atitude do Pessoal de Saúde , Monitorização Fisiológica/métodos , Monitorização Fisiológica/instrumentação , Pessoa de Meia-Idade , Cuidados Críticos/métodos
2.
Int J Med Inform ; 184: 105349, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38301520

RESUMO

BACKGROUND: Alarm fatigue is a major technology-induced hazard for patients and staff in intensive care units. Too many - mostly unnecessary - alarms cause desensitisation and lack of response in medical staff. Unsuitable alarm policies are one reason for alarm fatigue. But changing alarm policies is a delicate issue since it concerns patient safety. OBJECTIVE: We present ARTEMIS, a novel, computer-aided clinical decision support system for policy makers that can help to considerably improve alarm policies using data from hospital information systems. METHODS: Policy makers can use different policy components from ARTEMIS' internal library to assemble tailor-made alarm policies for their intensive care units. Alternatively, policy makers can provide even more highly customised policy components as Python functions using data the hospital information systems. This can even include machine learning models - for example for setting alarm thresholds. Finally, policy makers can evaluate their system of policies and compare the resulting alarm loads. RESULTS: ARTEMIS reports and compares numbers of alarms caused by different alarm policies for an easily adaptable target population. ARTEMIS can compare policies side-by-side and provides grid comparisons and heat maps for parameter optimisation. For example, we found that the utility of alarm delays varies based on target population. Furthermore, policy makers can introduce virtual parameters that are not in the original data by providing a formula to compute them. Virtual parameters help measuring and alarming on the right metric, even if the patient monitors do not directly measure this metric. CONCLUSION: ARTEMIS does not release the policy maker from assessing the policy from a medical standpoint. But as a knowledge discovery and clinical decision support system, it provides a strong quantitative foundation for medical decisions. At comparatively low cost of implementation, ARTEMIS can have a substantial impact on patients and staff alike - with organisational, economic, and clinical benefits for the implementing hospital.


Assuntos
Fadiga de Alarmes do Pessoal de Saúde , Alarmes Clínicos , Humanos , Unidades de Terapia Intensiva , Monitorização Fisiológica/métodos , Políticas
3.
JMIR Med Inform ; 11: e43847, 2023 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-36943344

RESUMO

BACKGROUND: Increasing digitalization in the medical domain gives rise to large amounts of health care data, which has the potential to expand clinical knowledge and transform patient care if leveraged through artificial intelligence (AI). Yet, big data and AI oftentimes cannot unlock their full potential at scale, owing to nonstandardized data formats, lack of technical and semantic data interoperability, and limited cooperation between stakeholders in the health care system. Despite the existence of standardized data formats for the medical domain, such as Fast Healthcare Interoperability Resources (FHIR), their prevalence and usability for AI remain limited. OBJECTIVE: In this paper, we developed a data harmonization pipeline (DHP) for clinical data sets relying on the common FHIR data standard. METHODS: We validated the performance and usability of our FHIR-DHP with data from the Medical Information Mart for Intensive Care IV database. RESULTS: We present the FHIR-DHP workflow in respect of the transformation of "raw" hospital records into a harmonized, AI-friendly data representation. The pipeline consists of the following 5 key preprocessing steps: querying of data from hospital database, FHIR mapping, syntactic validation, transfer of harmonized data into the patient-model database, and export of data in an AI-friendly format for further medical applications. A detailed example of FHIR-DHP execution was presented for clinical diagnoses records. CONCLUSIONS: Our approach enables the scalable and needs-driven data modeling of large and heterogenous clinical data sets. The FHIR-DHP is a pivotal step toward increasing cooperation, interoperability, and quality of patient care in the clinical routine and for medical research.

4.
Sci Rep ; 12(1): 21801, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36526892

RESUMO

Intensive care units (ICU) are often overflooded with alarms from monitoring devices which constitutes a hazard to both staff and patients. To date, the suggested solutions to excessive monitoring alarms have remained on a research level. We aimed to identify patient characteristics that affect the ICU alarm rate with the goal of proposing a straightforward solution that can easily be implemented in ICUs. Alarm logs from eight adult ICUs of a tertiary care university-hospital in Berlin, Germany were retrospectively collected between September 2019 and March 2021. Adult patients admitted to the ICU with at least 24 h of continuous alarm logs were included in the study. The sum of alarms per patient per day was calculated. The median was 119. A total of 26,890 observations from 3205 patients were included. 23 variables were extracted from patients' electronic health records (EHR) and a multivariable logistic regression was performed to evaluate the association of patient characteristics and alarm rates. Invasive blood pressure monitoring (adjusted odds ratio (aOR) 4.68, 95%CI 4.15-5.29, p < 0.001), invasive mechanical ventilation (aOR 1.24, 95%CI 1.16-1.32, p < 0.001), heart failure (aOR 1.26, 95%CI 1.19-1.35, p < 0.001), chronic renal failure (aOR 1.18, 95%CI 1.10-1.27, p < 0.001), hypertension (aOR 1.19, 95%CI 1.13-1.26, p < 0.001), high RASS (aOR 1.22, 95%CI 1.18-1.25, p < 0.001) and scheduled surgical admission (aOR 1.22, 95%CI 1.13-1.32, p < 0.001) were significantly associated with a high alarm rate. Our study suggests that patient-specific alarm management should be integrated in the clinical routine of ICUs. To reduce the overall alarm load, particular attention regarding alarm management should be paid to patients with invasive blood pressure monitoring, invasive mechanical ventilation, heart failure, chronic renal failure, hypertension, high RASS or scheduled surgical admission since they are more likely to have a high contribution to noise pollution, alarm fatigue and hence compromised patient safety in ICUs.


Assuntos
Alarmes Clínicos , Insuficiência Cardíaca , Hipertensão , Falência Renal Crônica , Adulto , Humanos , Estudos Retrospectivos , Unidades de Terapia Intensiva , Monitorização Fisiológica
5.
Stud Health Technol Inform ; 294: 273-274, 2022 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-35612072

RESUMO

Alarms help to detect medical conditions in intensive care units and improve patient safety. However, up to 99% of alarms are non-actionable, i.e. alarm that did not trigger a medical intervention in a defined time frame. Reducing their amount through machine learning (ML) is hypothesized to be a promising approach to improve patient monitoring and alarm management. This retrospective study presents the technical and medical pre-processing steps to annotate alarms into actionable and non-actionable, creating a basis for ML applications.


Assuntos
Alarmes Clínicos , Cuidados Críticos , Humanos , Unidades de Terapia Intensiva , Aprendizado de Máquina , Monitorização Fisiológica , Estudos Retrospectivos
6.
Stud Health Technol Inform ; 294: 559-560, 2022 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-35612143

RESUMO

Routinely collected electronic health records (EHR) in clinical information systems (CIS) are often heterogeneous, have inconsistent data formats and lack of documentation. We use the well-known open-source database schema of MIMIC-IV to address this issue aiming to support collaborative secondary analysis. Over 154 million data records from a German ICU have already been mapped and inserted into the schema successfully. However, discrepancies between the German and US health systems as well as specifics in our clinical source data hinder the direct translation to MIMIC. Evaluating and improving mapping completeness is part of the ongoing research.


Assuntos
Documentação , Registros Eletrônicos de Saúde , Bases de Dados Factuais
8.
Aliment Pharmacol Ther ; 54(9): 1150-1161, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34570916

RESUMO

BACKGROUND: Glucagon-like peptide-1 receptor agonists may be a treatment option in patients with non-alcoholic fatty liver disease (NAFLD). AIMS: To investigate the effects of semaglutide on liver stiffness and liver fat in subjects with NAFLD using non-invasive magnetic resonance imaging (MRI) methods. METHODS: This randomised, double-blind, placebo-controlled trial enrolled subjects with liver stiffness 2.50-4.63 kPa by magnetic resonance elastography (MRE) and liver steatosis ≥10% by MRI proton density fat fraction (MRI-PDFF). The primary endpoint was change from baseline to week 48 in liver stiffness assessed by MRE. RESULTS: Sixty-seven subjects were randomised to once-daily subcutaneous semaglutide 0.4 mg (n = 34) or placebo (n = 33). Change from baseline in liver stiffness was not significantly different between semaglutide and placebo at week 48 (estimated treatment ratio 0.96 (95% CI 0.89, 1.03; P = 0.2798); significant differences in liver stiffness were not observed at weeks 24 or 72. Reductions in liver steatosis were significantly greater with semaglutide (estimated treatment ratios: 0.70 [0.59, 0.84], P = 0.0002; 0.47 [0.36, 0.60], P < 0.0001; and 0.50 [0.39, 0.66], P < 0.0001) and more subjects achieved a ≥ 30% reduction in liver fat content with semaglutide at weeks 24, 48 and 72, (all P < 0.001). Decreases in liver enzymes, body weight and HbA1c were also observed with semaglutide. CONCLUSIONS: The change in liver stiffness in subjects with NAFLD was not significantly different between semaglutide and placebo. However, semaglutide significantly reduced liver steatosis compared with placebo which, together with improvements in liver enzymes and metabolic parameters, suggests a positive impact on disease activity and metabolic profile. ClinicalTrials.gov identifier: NCT03357380.


Assuntos
Diabetes Mellitus Tipo 2 , Técnicas de Imagem por Elasticidade , Hepatopatia Gordurosa não Alcoólica , Método Duplo-Cego , Peptídeos Semelhantes ao Glucagon/uso terapêutico , Humanos , Imageamento por Ressonância Magnética , Hepatopatia Gordurosa não Alcoólica/diagnóstico por imagem , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico
9.
Adv Ther ; 37(12): 4866-4876, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33006125

RESUMO

INTRODUCTION: Non-alcoholic fatty liver disease (NAFLD), one of the most common liver diseases, is associated with liver-related complications and metabolic comorbidities. The phenotype is wide, ranging from simple steatosis to non-alcoholic steatohepatitis with advanced fibrosis. In this analysis of a phase 1 trial, clinical characteristics of screened subjects with NAFLD were studied according to the extent of fibrosis assessed using magnetic resonance elastography (MRE). METHODS: One hundred ninety-four subjects with body mass index (BMI) of 25-40 kg/m2 and suspected NAFLD were assessed by MRE and grouped by MRE thresholds as a proxy for fibrosis staging (groups 0-4). Data were summarized by group levels, and correlation analyses between MRE values and clinical parameters (including magnetic resonance imaging-proton density fat fraction) were performed. RESULTS: Most subjects had MRE values in the lower range (groups 0-1; N = 148). Type 2 diabetes (T2D) and BMI > 35 kg/m2 were more frequent in groups with higher than lower MRE values. Subjects in the highest MRE groups also tended to be older and have higher liver enzyme concentrations compared with lower MRE groups. No, or weak, correlations were found between MRE values and clinical parameters (all r values ≤ 0.45). CONCLUSIONS: There was considerable variation and overlap in clinical characteristics across the spectrum of liver stiffness. Although groups with high MRE values generally included more subjects with T2D and obesity, and had higher age and concentrations of liver enzymes, the clinical characteristics did not strongly correlate with MRE scores in this population. TRIAL REGISTRATION: Registered on Clinicaltrials.gov on November 29, 2017 (NCT03357380).


Assuntos
Diabetes Mellitus Tipo 2/complicações , Cirrose Hepática/diagnóstico por imagem , Hepatopatia Gordurosa não Alcoólica/diagnóstico por imagem , Obesidade/complicações , Adulto , Índice de Massa Corporal , Técnicas de Imagem por Elasticidade , Feminino , Humanos , Cirrose Hepática/complicações , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/complicações
10.
Diabetes Ther ; 10(2): 649-662, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30788808

RESUMO

INTRODUCTION: The aim of the present analysis was to characterise the absorption, distribution and elimination of semaglutide by means of population pharmacokinetic (PK) models using data from nine clinical pharmacology trials conducted in both healthy subjects and those with type 2 diabetes. METHODS: Data were obtained from trials with subcutaneous and intravenous administration of semaglutide that utilised frequent PK sampling and included a total of 353 subjects with 10,573 concentration values. RESULTS: Semaglutide PK properties across trials, drug product strengths and populations were well characterised by a two-compartment model with first-order absorption and elimination. For a typical subject with type 2 diabetes, clearance was estimated to be 0.0348 L/h [95% confidence interval (CI) 0.0327-0.0369 L/h], and the central and peripheral volumes were estimated to be 3.59 L (95% CI 3.28-3.90 L) and 4.10 L (95% CI 3.78-4.42 L), respectively (i.e. a total volume of distribution of 7.7 L). Interindividual variation was low (~ 15%) for both clearance and volumes of distribution, with low residual error (< 5%). Clearance and the total volume of distribution were approximately proportional to body weight. Minor differences were identified between healthy subjects and subjects with type 2 diabetes with respect to clearance and absorption rate, and between injection sites with respect to bioavailability. CONCLUSIONS: A novel two-compartment model was developed to provide the general characteristics of semaglutide absorption following subcutaneous administration, and of distribution and elimination across administration routes. Semaglutide PK was shown to be predictable across populations and administration routes and within subjects, and was primarily influenced by body weight. FUNDING: Novo Nordisk, Bagsværd, Denmark.

11.
Diabetes Ther ; 9(4): 1533-1547, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29907893

RESUMO

INTRODUCTION: Semaglutide, a new treatment option approved for the treatment of patients with type 2 diabetes mellitus, is a glucagon-like peptide-1 receptor agonist to be injected subcutaneously once weekly. This analysis used a population pharmacokinetic model of semaglutide to identify clinically relevant covariates for exposure. METHODS: A total of 1612 patients with up to seven pharmacokinetic observations each were included in the analysis. All subjects had type 2 diabetes mellitus and were enrolled in one of five trials in the phase III development program for subcutaneous semaglutide once weekly (the SUSTAIN program). The treatment duration of the trials varied from 30 to 104 weeks. RESULTS: No clinically relevant effects on the exposure were seen for sex, age, race, ethnicity, renal function, or injection site used, and semaglutide exposure was stable over time. Of the covariates chosen, only body weight had a relevant effect on the exposure of semaglutide. Few subjects developed semaglutide antibodies, and the antibodies had no effect on exposure. Dose proportionality was shown for the 0.5 mg and 1.0 mg maintenance doses of semaglutide. CONCLUSION: The population pharmacokinetic study showed that semaglutide exposure is not affected by covariates other than body weight at either a maintenance dose of 0.5 or 1.0 mg semaglutide. Therefore, we conclude that no semaglutide dose adjustments are needed in different populations. This finding is to be further explored in an exposure-response analysis. TRIAL REGISTRATION: The trials were registered at ClinicalTrials.gov (identifiers: NCT02054897, NCT01930188, NCT01885208, NCT01720446 and NCT02207374). FUNDING: Novo Nordisk A/S, Bagsværd, Denmark.

12.
Diabetes Ther ; 9(4): 1441-1456, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29799100

RESUMO

INTRODUCTION: Semaglutide is a glucagon-like peptide-1 (GLP-1) analog approved for the once-weekly treatment of type 2 diabetes. The objective of this 16-week, double-blind, single-center thorough QT study was to confirm that semaglutide treatment does not prolong cardiac repolarization versus placebo. Prolongation of the QT interval is a biomarker for ventricular tachyarrhythmia. METHODS: In a parallel design, 168 healthy subjects were randomized to the treatment or placebo arms, of whom 166 were treated with subcutaneous semaglutide (N = 83; escalated to a supratherapeutic dose of 1.5 mg) or placebo (N = 83). The subjects (60% males) had a mean age of 38.2 years and body mass index of 25.1 kg/m2. To assess QT assay sensitivity, subjects in the placebo group received a single 400 mg moxifloxacin dose as positive control, and placebo in a crossover fashion. The primary endpoint was the time-matched change from baseline in QT interval corrected individually for heart rate (ΔQTcI), calculated from 11 electrocardiogram recordings from 0 to 48 h after the last 1.5 mg dose. Similar assessments were made for the therapeutic 0.5 and 1.0 mg semaglutide dose levels. RESULTS: No QTcI prolongation occurred with any semaglutide dose; the upper limits of two-sided 90% confidence intervals of the placebo-subtracted ΔQTcI were < 10 ms at all doses and time points. Exposure-response analysis showed no dependence of QTcI on semaglutide concentration. QT assay sensitivity was confirmed. The semaglutide safety profile was similar to that of other GLP-1 receptor agonists. CONCLUSION: Based on investigations of QT/QTc, no concern with regard to ventricular arrhythmias was raised as semaglutide did not prolong the cardiac repolarization duration in healthy subjects. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT 02064348. FUNDING: Novo Nordisk.

13.
Diabetes Obes Metab ; 20(9): 2238-2245, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29748996

RESUMO

AIMS: To evaluate dose levels for semaglutide, a glucagon-like peptide-1 analogue approved for the treatment of type 2 diabetes, by examining the effects of demographic factors on efficacy and safety in an exposure-response analysis. METHODS: We analysed data from 1552 adults from four randomized phase III trials of 30 to 56 weeks' duration, investigating once-weekly semaglutide doses 0.5 and 1.0 mg. Exposure-response relationships were investigated using graphical and model-based techniques to assess the two dose levels and subgroups with the highest and lowest exposure and response. RESULTS: The population had the following demographic characteristics: baseline mean age between 53.2 and 58.4 years, glycated haemoglobin (HbA1c) between 64 and 67 mmol/mol (8.0% and 8.3%), body weight between 71.3 and 96.2 kg, and diabetes duration between 4.2 and 8.9 years. Exposure-response analysis showed a clear HbA1c and weight reduction across exposures after 30 weeks, irrespective of baseline values. The exposure-response for HbA1c was influenced by baseline HbA1c, and body weight exposure-response was influenced by sex, with limited impact of other factors. Analyses for relevant subgroups of baseline body weight, baseline HbA1c and sex indicated clinically relevant additional benefits with regard to HbA1c and weight with 1.0 vs 0.5 mg semaglutide. The proportion of participants reporting gastrointestinal (GI) side effects increased with increasing exposure, but was counteracted by tolerance development. CONCLUSIONS: The analysis showed that all subgroups obtained a clinically relevant benefit with semaglutide 0.5 mg and an additional benefit with semaglutide 1.0 mg. The increase in GI side effects with higher exposure was mitigated by gradually increasing the dose.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Peptídeos Semelhantes ao Glucagon/administração & dosagem , Hipoglicemiantes/administração & dosagem , Glicemia/efeitos dos fármacos , Ensaios Clínicos Fase III como Assunto , Diabetes Mellitus Tipo 2/sangue , Relação Dose-Resposta a Droga , Feminino , Hemoglobinas Glicadas/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento , Redução de Peso/efeitos dos fármacos
14.
Adv Ther ; 35(4): 531-544, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29536338

RESUMO

INTRODUCTION: Semaglutide is a glucagon-like peptide-1 analogue for once-weekly subcutaneous treatment of type 2 diabetes. This trial compared the pharmacokinetics, pharmacodynamics, and safety of semaglutide in Japanese and Caucasian subjects. METHODS: In this single-center, double-blind, parallel-group, 13-week trial, 44 healthy male subjects (22 Japanese, 22 Caucasian) were randomized within each race to semaglutide 0.5 mg (n = 8), 1.0 mg (n = 8), placebo 0.5 mg (n = 3) or 1.0 mg (n = 3). The primary endpoint was semaglutide exposure at steady state [area under the curve (AUC0-168h)]. RESULTS: Steady-state exposure of semaglutide was similar for both populations: AUC0-168h estimated race ratio (ERR), Japanese/Caucasian: 0.5 mg, 1.06; 1.0 mg, 0.99; maximum concentration (Cmax) ERR: 0.5 mg, 1.06; 1.0 mg, 1.02. Exposure after the first dose (0.25 mg) was slightly higher in Japanese versus Caucasian subjects (AUC0-168h ERR 1.11; Cmax ERR 1.14). Dose-dependent increases in AUC0-168h and Cmax occurred in both populations. Accumulation was as expected, based on the half-life (t1/2, ~ 1 week) and dosing interval of semaglutide. Significant body weight reductions were observed with semaglutide 0.5 mg and 1.0 mg in Japanese (both p ≤ 0.05) and Caucasian (both p ≤ 0.05) subjects versus placebo. No new safety issues were identified. CONCLUSIONS: The pharmacokinetic, pharmacodynamic, and safety profiles of semaglutide were similar in Japanese and Caucasian subjects, suggesting that no dose adjustment is required for the clinical use of semaglutide in Japanese subjects. FUNDING: Novo Nordisk A/S, Denmark. TRIAL REGISTRATION: ClinicalTrials.gov identifier NCT02146079. Japanese trial registration number JapicCTI-142550.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Peptídeos Semelhantes ao Glucagon/farmacologia , Peptídeos Semelhantes ao Glucagon/uso terapêutico , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Adulto , Área Sob a Curva , Povo Asiático , Peso Corporal , Diabetes Mellitus Tipo 2/etnologia , Método Duplo-Cego , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Peptídeos Semelhantes ao Glucagon/farmacocinética , Meia-Vida , Humanos , Hipoglicemiantes/farmacocinética , Injeções Subcutâneas , Masculino , Pessoa de Meia-Idade , Redução de Peso , População Branca , Adulto Jovem
15.
Diabetes Obes Metab ; 20(3): 610-619, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28941314

RESUMO

AIM: To investigate the effects of semaglutide on fasting and postprandial glucose and lipid responses, and on gastric emptying. MATERIALS AND METHODS: This was a randomized, double-blind, placebo-controlled, 2-period, crossover trial. Subjects with obesity (N = 30) received once-weekly subcutaneous semaglutide, dose-escalated to 1.0 mg, or placebo. After each 12-week treatment period, glucose and lipid metabolism were assessed before and after standardized meals. Gastric emptying (paracetamol absorption test) and peptide YY (PYY) response were also assessed. RESULTS: Semaglutide treatment significantly lowered fasting concentrations of glucose and glucagon, and increased insulin vs placebo (estimated treatment ratio: 0.95 [95% confidence interval: 0.91, 0.98]; 0.86 [0.75, 0.98]; 1.45 [1.20, 1.75], respectively). Postprandial glucose metabolism significantly improved with semaglutide vs placebo (incremental area under the curve 0 to 5 hours [iAUC0-5h ]; estimated treatment difference: glucose -1.34 mmol h/L [-2.42, -0.27]; insulin -921 pmol h/L [-1461, -381]; C-peptide -1.42 nmol h/L [-2.33, -0.51]). Fasting and postprandial lipid metabolism improved with semaglutide vs placebo. First-hour gastric emptying after the meal was delayed with semaglutide vs placebo (AUC0-1h ; estimated treatment ratio: 0.73 [0.61, 0.87]); this may have contributed to the lower postprandial glucose increase in semaglutide-treated subjects. Overall gastric emptying (AUC0-5h ) was not statistically different between treatments. Fasting and postprandial PYY responses were significantly lower with semaglutide vs placebo (P = .0397 and P = .0097, respectively). CONCLUSION: Semaglutide improved fasting and postprandial glucose and lipid metabolism. Overall gastric emptying was similar to that with placebo; however, the observed first-hour delay with semaglutide may contribute to a slower entry of glucose into the circulation.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Esvaziamento Gástrico/efeitos dos fármacos , Peptídeos Semelhantes ao Glucagon/administração & dosagem , Hipoglicemiantes/administração & dosagem , Obesidade/complicações , Adulto , Glicemia/metabolismo , Estudos Cross-Over , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/fisiopatologia , Método Duplo-Cego , Esquema de Medicação , Jejum/sangue , Feminino , Humanos , Injeções Subcutâneas , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Obesidade/fisiopatologia , Período Pós-Prandial
16.
Diabetologia ; 60(8): 1390-1399, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28526920

RESUMO

AIMS/HYPOTHESIS: Semaglutide is a glucagon-like peptide-1 analogue in development for the treatment of type 2 diabetes. Its effects on first- and second-phase insulin secretion and other measures of beta cell function and glycaemic control were assessed. METHODS: In this single-centre, double-blind, placebo-controlled, parallel-group trial, conducted at the Profil Institut für Stoffwechselforschung, Germany, 75 adult (aged 18-64 years) participants with type 2 diabetes (eligibility: HbA1c of 6.5-9.0% (47.5-74.9 mmol/mol); BMI 20.0-35.0 kg/m2; and treatment with diet and exercise and/or metformin monotherapy with a dose unchanged in the 30 days prior to screening) were randomised (1:1) to once-weekly s.c. semaglutide 1.0 mg (0.25, 0.5, 1.0 mg escalated) or placebo for 12 weeks. Co-primary endpoints were changes from baseline to end of treatment in the first (AUC0-10 min) and second (AUC10-120 min) insulin secretion phases, as measured by the IVGTT. An arginine stimulation test (AST) and a 24 h meal stimulation test were also conducted. A graded glucose infusion test (GGIT) assessed insulin secretion rate (ISR) in treated participants and a group of untreated healthy participants. Safety endpoints were also assessed. RESULTS: In total, 37 participants received semaglutide and 38 received placebo. Following IVGTT, for insulin, both AUC0-10min and AUC10-120min were significantly increased with semaglutide (estimated treatment ratio [95% CI] 3.02 [2.53, 3.60] and 2.10 [1.86, 2.37], respectively; p < 0.0001). The 24 h meal test showed reduced fasting, postprandial and overall (AUC0-24h) glucose and glucagon responses with semaglutide (p < 0.0001). The AST showed that maximal insulin capacity increased following semaglutide treatment. During GGIT, semaglutide significantly increased ISR to levels similar to those in healthy participants. Semaglutide was well tolerated. CONCLUSIONS/INTERPRETATION: Twelve weeks of once-weekly treatment with semaglutide significantly improved beta cell function and glycaemic control in participants with type 2 diabetes. TRIAL REGISTRATION: ClinicalTrials.gov NCT02212067 FUNDING: The study was funded by Novo Nordisk A/S.


Assuntos
Glicemia/efeitos dos fármacos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Peptídeos Semelhantes ao Glucagon/uso terapêutico , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Adolescente , Adulto , Diabetes Mellitus Tipo 2/sangue , Método Duplo-Cego , Feminino , Peptídeos Semelhantes ao Glucagon/administração & dosagem , Hemoglobinas Glicadas/metabolismo , Humanos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/uso terapêutico , Metformina/administração & dosagem , Metformina/uso terapêutico , Pessoa de Meia-Idade , Gravidez , Adulto Jovem
17.
Diabetes Obes Metab ; 19(9): 1242-1251, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28266779

RESUMO

AIM: The aim of this trial was to investigate the mechanism of action for body weight loss with semaglutide. MATERIALS AND METHODS: This randomised, double-blind, placebo-controlled, two-period crossover trial investigated the effects of 12 weeks of treatment with once-weekly subcutaneous semaglutide, dose-escalated to 1.0 mg, in 30 subjects with obesity. Ad libitum energy intake, ratings of appetite, thirst, nausea and well-being, control of eating, food preference, resting metabolic rate, body weight and body composition were assessed. RESULTS: After a standardised breakfast, semaglutide, compared with placebo, led to a lower ad libitum energy intake during lunch (-1255 kJ; P < .0001) and during the subsequent evening meal ( P = .0401) and snacks ( P = .0034), resulting in a 24% reduction in total energy intake across all ad libitum meals throughout the day (-3036 kJ; P < .0001). Fasting overall appetite suppression scores were improved with semaglutide vs placebo, while nausea ratings were similar. Semaglutide was associated with less hunger and food cravings, better control of eating and a lower preference for high-fat foods. Resting metabolic rate, adjusted for lean body mass, did not differ between treatments. Semaglutide led to a reduction from baseline in mean body weight of 5.0 kg, predominantly from body fat mass. CONCLUSION: After 12 weeks of treatment, ad libitum energy intake was substantially lower with semaglutide vs placebo with a corresponding loss of body weight observed with semaglutide. In addition to reduced energy intake, likely mechanisms for semaglutide-induced weight loss included less appetite and food cravings, better control of eating and lower relative preference for fatty, energy-dense foods.


Assuntos
Adiposidade/efeitos dos fármacos , Depressores do Apetite/uso terapêutico , Regulação do Apetite/efeitos dos fármacos , Ingestão de Energia/efeitos dos fármacos , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Peptídeos Semelhantes ao Glucagon/uso terapêutico , Obesidade/tratamento farmacológico , Adulto , Depressores do Apetite/administração & dosagem , Depressores do Apetite/efeitos adversos , Metabolismo Basal/efeitos dos fármacos , Índice de Massa Corporal , Estudos Cross-Over , Relação Dose-Resposta a Droga , Método Duplo-Cego , Esquema de Medicação , Comportamento Alimentar/efeitos dos fármacos , Feminino , Preferências Alimentares/efeitos dos fármacos , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Peptídeos Semelhantes ao Glucagon/administração & dosagem , Peptídeos Semelhantes ao Glucagon/efeitos adversos , Humanos , Injeções Subcutâneas , Masculino , Pessoa de Meia-Idade , Obesidade/metabolismo , Autorrelato , Redução de Peso/efeitos dos fármacos
18.
Clin Pharmacokinet ; 56(11): 1381-1390, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28349386

RESUMO

BACKGROUND: The pharmacokinetics and tolerability of semaglutide, a once-weekly human glucagon-like peptide-1 analog in development for the treatment of type 2 diabetes mellitus, were investigated in subjects with/without renal impairment (RI). METHODS: Fifty-six subjects, categorized into renal function groups [normal, mild, moderate, severe, and end-stage renal disease (ESRD)], received a single subcutaneous dose of semaglutide 0.5 mg. Semaglutide plasma concentrations were assessed ≤480 h post-dose; the primary endpoint was the area under the plasma concentration-time curve from time zero to infinity. RESULTS: Semaglutide exposure in subjects with mild/moderate RI and ESRD was similar to that in subjects with normal renal function. In subjects with severe RI, the mean exposure of semaglutide was 22% higher than in subjects with normal renal function, and the 95% confidence interval (1.02-1.47) for the ratio exceeded the pre-specified limits (0.70-1.43). When adjusted for differences in sex, age, and body weight between the groups, all comparisons were within the pre-specified clinically relevant limits. Across RI groups there was no relationship between creatinine clearance (CLCR) and semaglutide exposure, or between CLCR and semaglutide maximum plasma drug concentration (C max). Hemodialysis did not appear to affect the pharmacokinetics of semaglutide. No appreciable changes in safety parameters or vital signs and no serious adverse events were noted. One subject with severe RI reported two major hypoglycemic events. CONCLUSION: When adjusted for differences in sex, age, and body weight, semaglutide exposure was similar between subjects with RI and subjects with normal renal function. Semaglutide (0.5 mg) was well-tolerated. Dose adjustment may not be warranted for subjects with RI. CLINICALTRIALS. GOV IDENTIFIER: NCT00833716.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Peptídeos Semelhantes ao Glucagon/efeitos adversos , Peptídeos Semelhantes ao Glucagon/farmacocinética , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/farmacocinética , Insuficiência Renal/complicações , Adolescente , Adulto , Idoso , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Feminino , Peptídeos Semelhantes ao Glucagon/administração & dosagem , Peptídeos Semelhantes ao Glucagon/sangue , Humanos , Hipoglicemiantes/sangue , Masculino , Pessoa de Meia-Idade , Insuficiência Renal/sangue , Adulto Jovem
19.
Clin Pharmacokinet ; 55(6): 657-72, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26597252

RESUMO

Liraglutide is an acylated glucagon-like peptide-1 analogue with 97 % amino acid homology with native glucagon-like peptide-1 and greatly protracted action. It is widely used for the treatment of type 2 diabetes mellitus, and administered by subcutaneous injection once daily. The pharmacokinetic properties of liraglutide enable 24-h exposure coverage, a requirement for 24-h glycaemic control with once-daily dosing. The mechanism of protraction relates to slowed release from the injection site, and a reduced elimination rate owing to metabolic stabilisation and reduced renal filtration. Drug exposure is largely independent of injection site, as well as age, race and ethnicity. Increasing body weight and male sex are associated with reduced concentrations, but there is substantial overlap between subgroups; therefore, dose escalation should be based on individual treatment outcome. Exposure is reduced with mild, moderate or severe renal or hepatic impairment. There are no clinically relevant changes in overall concentrations of various drugs (e.g. paracetamol, atorvastatin, griseofulvin, digoxin, lisinopril and oral combination contraceptives) when co-administered with liraglutide. Pharmacodynamic studies show multiple beneficial actions with liraglutide, including improved fasting and postprandial glycaemic control (mediated by increased insulin and reduced glucagon levels and minor delays in gastric emptying), reduced appetite and energy intake, and effects on postprandial lipid profiles. The counter-regulatory hormone response to hypoglycaemia is largely unaltered. The effects of liraglutide on insulin and glucagon secretion are glucose dependent, and hence the risk of hypoglycaemia is low. The pharmacokinetic and pharmacodynamic properties of liraglutide make it an important treatment option for many patients with type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacocinética , Hipoglicemiantes/uso terapêutico , Liraglutida/farmacocinética , Liraglutida/uso terapêutico , Área Sob a Curva , Glicemia , Peso Corporal , Interações Medicamentosas , Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Humanos , Hipoglicemiantes/farmacologia , Injeções Subcutâneas , Insulina/metabolismo , Lipídeos/sangue , Liraglutida/farmacologia , Falência Hepática/metabolismo , Insuficiência Renal/metabolismo , Fatores Sexuais
20.
J Clin Pharmacol ; 55(5): 497-504, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25475122

RESUMO

The effect of semaglutide, a once-weekly human glucagon-like peptide-1 (GLP-1) analog in development for type 2 diabetes (T2D), on the bioavailability of a combined oral contraceptive was investigated. Postmenopausal women with T2D (n = 43) on diet/exercise ± metformin received ethinylestradiol (0.03 mg)/levonorgestrel (0.15 mg) once daily for 8 days before (semaglutide-free) and during (steady-state 1.0 mg) semaglutide treatment (subcutaneous once weekly; dose escalation: 0.25 mg 4 weeks; 0.5 mg 4 weeks; 1.0 mg 5 weeks). Bioequivalence of oral contraceptives was established if 90%CI for the ratio of pharmacokinetic parameters during semaglutide steady-state and semaglutide-free periods was within prespecified limits (0.80-1.25). The bioequivalence criterion was met for ethinylestradiol area under the curve (AUC0-24 h ) for semaglutide steady-state/semaglutide-free; 1.11 (1.06-1.15). AUC0-24 h was 20% higher for levonorgestrel at semaglutide steady-state vs. semaglutide-free (1.20 [1.15-1.26]). Cmax was within bioequivalence criterion for both contraceptives. Reductions (mean ± SD) in HbA1c (-1.1 ± 0.6%) and weight (-4.3 ± 3.1 kg) were observed. Semaglutide pharmacokinetics were compatible with once-weekly dosing; the semaglutide dose and dose-escalation regimen were well tolerated. Adverse events, mainly gastrointestinal, were mild to moderate in severity. Asymptomatic increases in mean amylase and lipase were observed. Three subjects had elevated alanine aminotransferase levels ≥3x the upper limit of normal during semaglutide/oral contraceptive coadministration, which were reported as adverse events, but resolved during follow-up. Semaglutide did not reduce the bioavailability of ethinylestradiol and levonorgestrel.


Assuntos
Anticoncepcionais Orais Combinados/farmacocinética , Etinilestradiol/farmacocinética , Peptídeos Semelhantes ao Glucagon/farmacologia , Hipoglicemiantes/farmacologia , Levanogestrel/farmacocinética , Idoso , Área Sob a Curva , Glicemia , Combinação de Medicamentos , Interações Medicamentosas , Feminino , Peptídeos Semelhantes ao Glucagon/sangue , Hemoglobinas Glicadas , Meia-Vida , Humanos , Hipoglicemiantes/sangue , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Pós-Menopausa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...