Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Lancet Digit Health ; 5(9): e607-e617, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37543512

RESUMO

BACKGROUND: Exercise can rapidly drop glucose in people with type 1 diabetes. Ubiquitous wearable fitness sensors are not integrated into automated insulin delivery (AID) systems. We hypothesised that an AID can automate insulin adjustments using real-time wearable fitness data to reduce hypoglycaemia during exercise and free-living conditions compared with an AID not automating use of fitness data. METHODS: Our study population comprised of individuals (aged 21-50 years) with type 1 diabetes from from the Harold Schnitzer Diabetes Health Center clinic at Oregon Health and Science University, OR, USA, who were enrolled into a 76 h single-centre, two-arm randomised (4-block randomisation), non-blinded crossover study to use (1) an AID that detects exercise, prompts the user, and shuts off insulin during exercise using an exercise-aware adaptive proportional derivative (exAPD) algorithm or (2) an AID that automates insulin adjustments using fitness data in real-time through an exercise-aware model predictive control (exMPC) algorithm. Both algorithms ran on iPancreas comprising commercial glucose sensors, insulin pumps, and smartwatches. Participants executed 1 week run-in on usual therapy followed by exAPD or exMPC for one 12 h primary in-clinic session involving meals, exercise, and activities of daily living, and 2 free-living out-patient days. Primary outcome was time below range (<3·9 mmol/L) during the primary in-clinic session. Secondary outcome measures included mean glucose and time in range (3·9-10 mmol/L). This trial is registered with ClinicalTrials.gov, NCT04771403. FINDINGS: Between April 13, 2021, and Oct 3, 2022, 27 participants (18 females) were enrolled into the study. There was no significant difference between exMPC (n=24) versus exAPD (n=22) in time below range (mean [SD] 1·3% [2·9] vs 2·5% [7·0]) or time in range (63·2% [23·9] vs 59·4% [23·1]) during the primary in-clinic session. In the 2 h period after start of in-clinic exercise, exMPC had significantly lower mean glucose (7·3 [1·6] vs 8·0 [1·7] mmol/L, p=0·023) and comparable time below range (1·4% [4·2] vs 4·9% [14·4]). Across the 76 h study, both algorithms achieved clinical time in range targets (71·2% [16] and 75·5% [11]) and time below range (1·0% [1·2] and 1·3% [2·2]), significantly lower than run-in period (2·4% [2·4], p=0·0004 vs exMPC; p=0·012 vs exAPD). No adverse events occurred. INTERPRETATION: AIDs can integrate exercise data from smartwatches to inform insulin dosing and limit hypoglycaemia while improving glucose outcomes. Future AID systems that integrate exercise metrics from wearable fitness sensors may help people living with type 1 diabetes exercise safely by limiting hypoglycaemia. FUNDING: JDRF Foundation and the Leona M and Harry B Helmsley Charitable Trust, National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases.


Assuntos
Diabetes Mellitus Tipo 1 , Hipoglicemia , Dispositivos Eletrônicos Vestíveis , Feminino , Humanos , Atividades Cotidianas , Inteligência Artificial , Estudos Cross-Over , Diabetes Mellitus Tipo 1/tratamento farmacológico , Glucose/uso terapêutico , Gastos em Saúde , Hipoglicemiantes/uso terapêutico , Insulina , Estados Unidos , Masculino
2.
Am J Physiol Endocrinol Metab ; 325(3): E192-E206, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37436961

RESUMO

Exercise can cause dangerous fluctuations in blood glucose in people living with type 1 diabetes (T1D). Aerobic exercise, for example, can cause acute hypoglycemia secondary to increased insulin-mediated and noninsulin-mediated glucose utilization. Less is known about how resistance exercise (RE) impacts glucose dynamics. Twenty-five people with T1D underwent three sessions of either moderate or high-intensity RE at three insulin infusion rates during a glucose tracer clamp. We calculated time-varying rates of endogenous glucose production (EGP) and glucose disposal (Rd) across all sessions and used linear regression and extrapolation to estimate insulin- and noninsulin-mediated components of glucose utilization. Blood glucose did not change on average during exercise. The area under the curve (AUC) for EGP increased by 1.04 mM during RE (95% CI: 0.65-1.43, P < 0.001) and decreased proportionally to insulin infusion rate (0.003 mM per percent above basal rate, 95% CI: 0.001-0.006, P = 0.003). The AUC for Rd rose by 1.26 mM during RE (95% CI: 0.41-2.10, P = 0.004) and increased proportionally with insulin infusion rate (0.04 mM per percent above basal rate, CI: 0.03-0.04, P < 0.001). No differences were observed between the moderate and high resistance groups. Noninsulin-mediated glucose utilization rose significantly during exercise before returning to baseline roughly 30-min postexercise. Insulin-mediated glucose utilization remained unchanged during exercise sessions. Circulating catecholamines and lactate rose during exercise despite relatively small changes observed in Rd. Results provide an explanation of why RE may pose a lower overall risk for hypoglycemia.NEW & NOTEWORTHY Aerobic exercise is known to cause decreases in blood glucose secondary to increased glucose utilization in people living with type 1 diabetes (T1D). However, less is known about how resistance-type exercise impacts glucose dynamics. Twenty-five participants with T1D performed in-clinic weight-bearing exercises under a glucose clamp. Mathematical modeling of infused glucose tracer allowed for quantification of the rate of hepatic glucose production as well as rates of insulin-mediated and noninsulin-mediated glucose uptake experienced during resistance exercise.


Assuntos
Diabetes Mellitus Tipo 1 , Hipoglicemia , Treinamento Resistido , Humanos , Glucose , Insulina , Glicemia , Exercício Físico , Ácido Láctico
3.
NPJ Digit Med ; 6(1): 39, 2023 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-36914699

RESUMO

We present a robust insulin delivery system that includes automated meal detection and carbohydrate content estimation using machine learning for meal insulin dosing called robust artificial pancreas (RAP). We conducted a randomized, single-center crossover trial to compare postprandial glucose control in the four hours following unannounced meals using a hybrid model predictive control (MPC) algorithm and the RAP system. The RAP system includes a neural network model to automatically detect meals and deliver a recommended meal insulin dose. The meal detection algorithm has a sensitivity of 83.3%, false discovery rate of 16.6%, and mean detection time of 25.9 minutes. While there is no significant difference in incremental area under the curve of glucose, RAP significantly reduces time above range (glucose >180 mg/dL) by 10.8% (P = 0.04) and trends toward increasing time in range (70-180 mg/dL) by 9.1% compared with MPC. Time below range (glucose <70 mg/dL) is not significantly different between RAP and MPC.

4.
iScience ; 25(3): 103888, 2022 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-35252806

RESUMO

Prevention of hypoglycemia (glucose <70 mg/dL) during aerobic exercise is a major challenge in type 1 diabetes. Providing predictions of glycemic changes during and following exercise can help people with type 1 diabetes avoid hypoglycemia. A unique dataset representing 320 days and 50,000 + time points of glycemic measurements was collected in adults with type 1 diabetes who participated in a 4-arm crossover study evaluating insulin-pump therapies, whereby each participant performed eight identically designed in-clinic exercise studies. We demonstrate that even under highly controlled conditions, there is considerable intra-participant and inter-participant variability in glucose outcomes during and following exercise. Participants with higher aerobic fitness exhibited significantly lower minimum glucose and steeper glucose declines during exercise. Adaptive, personalized machine learning (ML) algorithms were designed to predict exercise-related glucose changes. These algorithms achieved high accuracy in predicting the minimum glucose and hypoglycemia during and following exercise sessions, for all fitness levels.

5.
Am J Physiol Endocrinol Metab ; 320(3): E425-E437, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33356994

RESUMO

Aerobic exercise in type 1 diabetes (T1D) causes rapid increase in glucose utilization due to muscle work during exercise, followed by increased insulin sensitivity after exercise. Better understanding of these changes is necessary for models of exercise in T1D. Twenty-six individuals with T1D underwent three sessions at three insulin rates (100%, 150%, 300% of basal). After 3-h run-in, participants performed 45 min aerobic exercise (moderate or intense). We determined area under the curve for endogenous glucose production (AUCEGP) and rate of glucose disappearance (AUCRd) over 45 min from exercise start. A novel application of linear regression of Rd across the three insulin sessions allowed separation of insulin-mediated from non-insulin-mediated glucose uptake before, during, and after exercise. AUCRd increased 12.45 mmol/L (CI = 10.33-14.58, P < 0.001) and 13.13 mmol/L (CI = 11.01-15.26, P < 0.001) whereas AUCEGP increased 1.66 mmol/L (CI = 1.01-2.31, P < 0.001) and 3.46 mmol/L (CI = 2.81-4.11, P < 0.001) above baseline during moderate and intense exercise, respectively. AUCEGP increased during intense exercise by 2.14 mmol/L (CI = 0.91-3.37, P < 0.001) compared with moderate exercise. There was significant effect of insulin infusion rate on AUCRd equal to 0.06 mmol/L per % above basal rate (CI = 0.05-0.07, P < 0.001). Insulin-mediated glucose uptake rose during exercise and persisted hours afterward, whereas non-insulin-mediated effect was limited to the exercise period. To our knowledge, this method of isolating dynamic insulin- and non-insulin-mediated uptake has not been previously employed during exercise. These results will be useful in informing glucoregulatory models of T1D. The study has been registered at www.clinicaltrials.gov as NCT03090451.NEW & NOTEWORTHY Separating insulin and non-insulin glucose uptake dynamically during exercise in type 1 diabetes has not been done before. We use a multistep process, including a previously described linear regression method, over three insulin infusion sessions, to perform this separation and can graph these components before, during, and after exercise for the first time.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Exercício Físico/fisiologia , Glucose/farmacocinética , Insulina/fisiologia , Adolescente , Adulto , Glicemia/metabolismo , Feminino , Humanos , Hiperinsulinismo/metabolismo , Hipoglicemia/metabolismo , Insulina/administração & dosagem , Insulina/metabolismo , Resistência à Insulina/fisiologia , Masculino , Pessoa de Meia-Idade , Esforço Físico/fisiologia , Adulto Jovem
6.
Diabetes Care ; 43(11): 2721-2729, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32907828

RESUMO

OBJECTIVE: To assess the efficacy and feasibility of a dual-hormone (DH) closed-loop system with insulin and a novel liquid stable glucagon formulation compared with an insulin-only closed-loop system and a predictive low glucose suspend (PLGS) system. RESEARCH DESIGN AND METHODS: In a 76-h, randomized, crossover, outpatient study, 23 participants with type 1 diabetes used three modes of the Oregon Artificial Pancreas system: 1) dual-hormone (DH) closed-loop control, 2) insulin-only single-hormone (SH) closed-loop control, and 3) PLGS system. The primary end point was percentage time in hypoglycemia (<70 mg/dL) from the start of in-clinic aerobic exercise (45 min at 60% VO2max) to 4 h after. RESULTS: DH reduced hypoglycemia compared with SH during and after exercise (DH 0.0% [interquartile range 0.0-4.2], SH 8.3% [0.0-12.5], P = 0.025). There was an increased time in hyperglycemia (>180 mg/dL) during and after exercise for DH versus SH (20.8% DH vs. 6.3% SH, P = 0.038). Mean glucose during the entire study duration was DH, 159.2; SH, 151.6; and PLGS, 163.6 mg/dL. Across the entire study duration, DH resulted in 7.5% more time in target range (70-180 mg/dL) compared with the PLGS system (71.0% vs. 63.4%, P = 0.044). For the entire study duration, DH had 28.2% time in hyperglycemia vs. 25.1% for SH (P = 0.044) and 34.7% for PLGS (P = 0.140). Four participants experienced nausea related to glucagon, leading three to withdraw from the study. CONCLUSIONS: The glucagon formulation demonstrated feasibility in a closed-loop system. The DH system reduced hypoglycemia during and after exercise, with some increase in hyperglycemia.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Glucagon/administração & dosagem , Sistemas de Infusão de Insulina , Insulina/administração & dosagem , Pâncreas Artificial , Adulto , Glicemia/análise , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Estudos Cross-Over , Diabetes Mellitus Tipo 1/sangue , Exercício Físico/fisiologia , Estudos de Viabilidade , Feminino , Glucagon/efeitos adversos , Humanos , Hiperglicemia/induzido quimicamente , Hiperglicemia/tratamento farmacológico , Hipoglicemia/induzido quimicamente , Hipoglicemia/tratamento farmacológico , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/efeitos adversos , Insulina/efeitos adversos , Masculino , Pessoa de Meia-Idade , Oregon , Pacientes Ambulatoriais , Adulto Jovem
7.
Nat Metab ; 2(7): 612-619, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32694787

RESUMO

Type 1 diabetes (T1D) is characterized by pancreatic beta cell dysfunction and insulin depletion. Over 40% of people with T1D manage their glucose through multiple injections of long-acting basal and short-acting bolus insulin, so-called multiple daily injections (MDI)1,2. Errors in dosing can lead to life-threatening hypoglycaemia events (<70 mg dl-1) and hyperglycaemia (>180 mg dl-1), increasing the risk of retinopathy, neuropathy, and nephropathy. Machine learning (artificial intelligence) approaches are being harnessed to incorporate decision support into many medical specialties. Here, we report an algorithm that provides weekly insulin dosage recommendations to adults with T1D using MDI therapy. We employ a unique virtual platform3 to generate over 50,000 glucose observations to train a k-nearest neighbours4 decision support system (KNN-DSS) to identify causes of hyperglycaemia or hypoglycaemia and determine necessary insulin adjustments from a set of 12 potential recommendations. The KNN-DSS algorithm achieves an overall agreement with board-certified endocrinologists of 67.9% when validated on real-world human data, and delivers safe recommendations, per endocrinologist review. A comparison of inter-physician-recommended adjustments to insulin pump therapy indicates full agreement of 41.2% among endocrinologists, which is consistent with previous measures of inter-physician agreement (41-45%)5. In silico3,6 benchmarking using a platform accepted by the United States Food and Drug Administration for evaluation of artificial pancreas technologies indicates substantial improvement in glycaemic outcomes after 12 weeks of KNN-DSS use. Our data indicate that the KNN-DSS allows for early identification of dangerous insulin regimens and may be used to improve glycaemic outcomes and prevent life-threatening complications in people with T1D.


Assuntos
Inteligência Artificial , Sistemas de Apoio a Decisões Clínicas , Diabetes Mellitus Tipo 1/tratamento farmacológico , Adulto , Algoritmos , Glicemia/análise , Simulação por Computador , Gerenciamento Clínico , Controle Glicêmico , Humanos , Hiperglicemia/sangue , Hipoglicemia/sangue , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/sangue , Hipoglicemiantes/uso terapêutico , Insulina/administração & dosagem , Insulina/sangue , Insulina/uso terapêutico , Sistemas de Infusão de Insulina , Reprodutibilidade dos Testes
8.
Diabetes Technol Ther ; 22(11): 801-811, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32297795

RESUMO

Background: Despite new glucose sensing technologies, nocturnal hypoglycemia is still a problem for people with type 1 diabetes (T1D) as symptoms and sensor alarms may not be detected while sleeping. Accurately predicting nocturnal hypoglycemia before sleep may help minimize nighttime hypoglycemia. Methods: A support vector regression (SVR) model was trained to predict, before bedtime, the overnight minimum glucose and overnight nocturnal hypoglycemia for people with T1D. The algorithm was trained on continuous glucose measurements and insulin data collected from 124 people (22,804 valid nights of data) with T1D. The minimum glucose threshold for announcing nocturnal hypoglycemia risk was derived by applying a decision theoretic criterion to maximize expected net benefit. Accuracy was evaluated on a validation set from 10 people with T1D during a 4-week trial under free-living sensor-augmented insulin-pump therapy. The primary outcome measures were sensitivity and specificity of prediction, the correlation between predicted and actual minimum nocturnal glucose, and root-mean-square error. The impact of using the algorithm to prevent nocturnal hypoglycemia is shown in-silico. Results: The algorithm predicted 94.1% of nocturnal hypoglycemia events (<3.9 mmol/L, 95% confidence interval [CI], 71.3-99.9) with an area under the receiver operating characteristic curve of 0.86 (95% CI, 0.75-0.98). Correlation between actual and predicted minimum glucose was high (R = 0.71, P < 0.001). In-silico simulations showed that the algorithm could reduce nocturnal hypoglycemia by 77.0% (P = 0.006) without impacting time in target range (3.9-10 mmol/L). Conclusion: An SVR model trained on a big data set and optimized using decision theoretic criterion can accurately predict at bedtime if overnight nocturnal hypoglycemia will occur and may help reduce nocturnal hypoglycemia.


Assuntos
Diabetes Mellitus Tipo 1 , Hipoglicemia , Adulto , Glicemia , Ciência de Dados , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/tratamento farmacológico , Feminino , Humanos , Hipoglicemia/diagnóstico , Hipoglicemia/prevenção & controle , Sistemas de Infusão de Insulina , Masculino , Sono , Tempo
9.
PLoS One ; 14(7): e0217301, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31344037

RESUMO

PURPOSE: We introduce two validated single (SH) and dual hormone (DH) mathematical models that represent an in-silico virtual patient population (VPP) for type 1 diabetes (T1D). The VPP can be used to evaluate automated insulin and glucagon delivery algorithms, so-called artificial pancreas (AP) algorithms that are currently being used to help people with T1D better manage their glucose levels. We present validation results comparing these virtual patients with true clinical patients undergoing AP control and demonstrate that the virtual patients behave similarly to people with T1D. METHODS: A single hormone virtual patient population (SH-VPP) was created that is comprised of eight differential equations that describe insulin kinetics, insulin dynamics and carbohydrate absorption. The parameters in this model that represent insulin sensitivity were statistically sampled from a normal distribution to create a population of virtual patients with different levels of insulin sensitivity. A dual hormone virtual patient population (DH-VPP) extended this SH-VPP by incorporating additional equations to represent glucagon kinetics and glucagon dynamics. The DH-VPP is comprised of thirteen differential equations and a parameter representing glucagon sensitivity, which was statistically sampled from a normal distribution to create virtual patients with different levels of glucagon sensitivity. We evaluated the SH-VPP and DH-VPP on a clinical data set of 20 people with T1D who participated in a 3.5-day outpatient AP study. Twenty virtual patients were matched with the 20 clinical patients by total daily insulin requirements and body weight. The identical meals given during the AP study were given to the virtual patients and the identical AP control algorithm that was used to control the glucose of the virtual patients was used on the clinical patients. We compared percent time in target range (70-180 mg/dL), time in hypoglycemia (<70 mg/dL) and time in hyperglycemia (>180 mg/dL) for both the virtual patients and the actual patients. RESULTS: The subjects in the SH-VPP performed similarly vs. the actual patients (time in range: 78.1 ± 5.1% vs. 74.3 ± 8.1%, p = 0.11; time in hypoglycemia: 3.4 ± 1.3% vs. 2.8 ± 1.7%, p = 0.23). The subjects in the DH-VPP also performed similarly vs. the actual patients (time in range: 75.6 ± 5.5% vs. 71.9 ± 10.9%, p = 0.13; time in hypoglycemia: 0.9 ± 0.8% vs. 1.3 ± 1%, p = 0.19). While the VPPs tended to over-estimate the time in range relative to actual patients, the difference was not statistically significant. CONCLUSIONS: We have verified that a SH-VPP and a DH-VPP performed comparably with actual patients undergoing AP control using an identical control algorithm. The SH-VPP and DH-VPP may be used as a simulator for pre-evaluation of T1D control algorithms.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Tipo 1 , Glucagon/sangue , Insulina , Modelos Biológicos , Adulto , Idoso , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/fisiopatologia , Feminino , Humanos , Insulina/sangue , Insulina/uso terapêutico , Cinética , Masculino , Pessoa de Meia-Idade
10.
J Diabetes Sci Technol ; 13(5): 919-927, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30650997

RESUMO

BACKGROUND: Fear of exercise related hypoglycemia is a major reason why people with type 1 diabetes (T1D) do not exercise. There is no validated prediction algorithm that can predict hypoglycemia at the start of aerobic exercise. METHODS: We have developed and evaluated two separate algorithms to predict hypoglycemia at the start of exercise. Model 1 is a decision tree and model 2 is a random forest model. Both models were trained using a meta-data set based on 154 observations of in-clinic aerobic exercise in 43 adults with T1D from 3 different studies that included participants using sensor augmented pump therapy, automated insulin delivery therapy, and automated insulin and glucagon therapy. Both models were validated using an entirely new validation data set with 90 exercise observations collected from 12 new adults with T1D. RESULTS: Model 1 identified two critical features predictive of hypoglycemia during exercise: heart rate and glucose at the start of exercise. If heart rate was greater than 121 bpm during the first 5 min of exercise and glucose at the start of exercise was less than 182 mg/dL, it predicted hypoglycemia with 79.55% accuracy. Model 2 achieved a higher accuracy of 86.7% using additional features and higher complexity. CONCLUSIONS: Models presented here can assist people with T1D to avoid exercise related hypoglycemia. The simple model 1 heuristic can be easily remembered (the 180/120 rule) and model 2 is more complex requiring computational resources, making it suitable for automated artificial pancreas or decision support systems.


Assuntos
Diabetes Mellitus Tipo 1/sangue , Exercício Físico/fisiologia , Hipoglicemia , Aprendizado de Máquina , Adulto , Glicemia , Diabetes Mellitus Tipo 1/tratamento farmacológico , Feminino , Frequência Cardíaca , Humanos , Hipoglicemia/sangue , Hipoglicemia/fisiopatologia , Masculino , Pâncreas Artificial
11.
Can J Diabetes ; 43(6): 406-414.e1, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30414785

RESUMO

OBJECTIVES: Physical exercise is recommended for individuals with type 1 diabetes, yet the effects of exercise on glycemic control are not well established. We evaluated the impact of different modes of exercise on glycemic control in people with type 1 diabetes. METHODS: In a 3-week randomized crossover trial, 10 adults with type 1 diabetes (4 men and 6 women, aged 33±6 years; duration of diabetes, 18±10 years; glycated hemoglobin level, 7.4%±1%) were assigned to 3 weeks of intervention: aerobic exercise (treadmill at 60% of maximum volume of oxygen utilization), resistance training (8 to 12 repetitions of 5 upper and lower body exercises at 60% to 80% of 1 repetition maximum) or no exercise (control). During each exercise week, participants completed 2 monitored 45 min exercise sessions. For each week of the study, we analyzed participants' insulin pump data, sensor glucose data and meal intake using a custom smart-phone application. The primary outcome was the percentage of time in range (glucose >3.9 mmol/L and ≤10 mmol/L) for the 24 h after each bout of exercise or rest during the control week. The study was registered on ClinicalTrials.gov (NCT:02687893). RESULTS: Aerobic exercise caused a mean glucose reduction during exercise of 3.94±2.67 mmol/L, whereas the reduction during resistance training was 1.33±1.78 mmol/L (p=0.007). The mean percentage time in range for the 24 h after resistance training was significantly greater than that during the control period (70% vs. 56%, p=0.013) but not after aerobic exercise (60%). CONCLUSIONS: The results indicate that when various confounders are considered, resistance training could improve glycemic control in this population.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Exercício Físico , Hiperglicemia/prevenção & controle , Hipoglicemia/prevenção & controle , Cooperação do Paciente/estatística & dados numéricos , Treinamento Resistido , Adulto , Estudos Cross-Over , Feminino , Seguimentos , Humanos , Masculino , Prognóstico
12.
JMIR Mhealth Uhealth ; 6(12): e10338, 2018 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-30530451

RESUMO

BACKGROUND: Wrist-worn activity monitors are often used to monitor heart rate (HR) and energy expenditure (EE) in a variety of settings including more recently in medical applications. The use of real-time physiological signals to inform medical systems including drug delivery systems and decision support systems will depend on the accuracy of the signals being measured, including accuracy of HR and EE. Prior studies assessed accuracy of wearables only during steady-state aerobic exercise. OBJECTIVE: The objective of this study was to validate the accuracy of both HR and EE for 2 common wrist-worn devices during a variety of dynamic activities that represent various physical activities associated with daily living including structured exercise. METHODS: We assessed the accuracy of both HR and EE for two common wrist-worn devices (Fitbit Charge 2 and Garmin vívosmart HR+) during dynamic activities. Over a 2-day period, 20 healthy adults (age: mean 27.5 [SD 6.0] years; body mass index: mean 22.5 [SD 2.3] kg/m2; 11 females) performed a maximal oxygen uptake test, free-weight resistance circuit, interval training session, and activities of daily living. Validity was assessed using an HR chest strap (Polar) and portable indirect calorimetry (Cosmed). Accuracy of the commercial wearables versus research-grade standards was determined using Bland-Altman analysis, correlational analysis, and error bias. RESULTS: Fitbit and Garmin were reasonably accurate at measuring HR but with an overall negative bias. There was more error observed during high-intensity activities when there was a lack of repetitive wrist motion and when the exercise mode indicator was not used. The Garmin estimated HR with a mean relative error (RE, %) of -3.3% (SD 16.7), whereas Fitbit estimated HR with an RE of -4.7% (SD 19.6) across all activities. The highest error was observed during high-intensity intervals on bike (Fitbit: -11.4% [SD 35.7]; Garmin: -14.3% [SD 20.5]) and lowest error during high-intensity intervals on treadmill (Fitbit: -1.7% [SD 11.5]; Garmin: -0.5% [SD 9.4]). Fitbit and Garmin EE estimates differed significantly, with Garmin having less negative bias (Fitbit: -19.3% [SD 28.9], Garmin: -1.6% [SD 30.6], P<.001) across all activities, and with both correlating poorly with indirect calorimetry measures. CONCLUSIONS: Two common wrist-worn devices (Fitbit Charge 2 and Garmin vívosmart HR+) show good HR accuracy, with a small negative bias, and reasonable EE estimates during low to moderate-intensity exercise and during a variety of common daily activities and exercise. Accuracy was compromised markedly when the activity indicator was not used on the watch or when activities involving less wrist motion such as cycle ergometry were done.

13.
Diabetes Care ; 41(7): 1471-1477, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29752345

RESUMO

OBJECTIVE: Automated insulin delivery is the new standard for type 1 diabetes, but exercise-related hypoglycemia remains a challenge. Our aim was to determine whether a dual-hormone closed-loop system using wearable sensors to detect exercise and adjust dosing to reduce exercise-related hypoglycemia would outperform other forms of closed-loop and open-loop therapy. RESEARCH DESIGN AND METHODS: Participants underwent four arms in randomized order: dual-hormone, single-hormone, predictive low glucose suspend, and continuation of current care over 4 outpatient days. Each arm included three moderate-intensity aerobic exercise sessions. The two primary outcomes were percentage of time in hypoglycemia (<70 mg/dL) and in a target range (70-180 mg/dL) assessed across the entire study and from the start of the in-clinic exercise until the next meal. RESULTS: The analysis included 20 adults with type 1 diabetes who completed all arms. The mean time (SD) in hypoglycemia was the lowest with dual-hormone during the exercise period: 3.4% (4.5) vs. 8.3% (12.6) single-hormone (P = 0.009) vs. 7.6% (8.0) predictive low glucose suspend (P < 0.001) vs. 4.3% (6.8) current care where pre-exercise insulin adjustments were allowed (P = 0.49). Time in hypoglycemia was also the lowest with dual-hormone during the entire 4-day study: 1.3% (1.0) vs. 2.8% (1.7) single-hormone (P < 0.001) vs. 2.0% (1.5) predictive low glucose suspend (P = 0.04) vs. 3.1% (3.2) current care (P = 0.007). Time in range during the entire study was the highest with single-hormone: 74.3% (8.0) vs. 72.0% (10.8) dual-hormone (P = 0.44). CONCLUSIONS: The addition of glucagon delivery to a closed-loop system with automated exercise detection reduces hypoglycemia in physically active adults with type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Exercício Físico/fisiologia , Glucagon/administração & dosagem , Hipoglicemiantes/administração & dosagem , Sistemas de Infusão de Insulina , Insulina/administração & dosagem , Dispositivos Eletrônicos Vestíveis , Adulto , Glicemia/análise , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Automonitorização da Glicemia/instrumentação , Automonitorização da Glicemia/métodos , Estudos Cross-Over , Diabetes Mellitus Tipo 1/sangue , Feminino , Humanos , Sistemas de Infusão de Insulina/normas , Masculino , Refeições , Pessoa de Meia-Idade , Pacientes Ambulatoriais , Pâncreas Artificial , Adulto Jovem
14.
Diabetes Obes Metab ; 20(2): 443-447, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28718987

RESUMO

The aim of this pilot study was to investigate the effect of exercise on sleep and nocturnal hypoglycaemia in adults with type 1 diabetes (T1D). In a 3-week crossover trial, 10 adults with T1D were randomized to perform aerobic, resistance or no exercise. During each exercise week, participants completed 2 separate 45-minutes exercise sessions at an academic medical center. Participants returned home and wore a continuous glucose monitor and a wrist-based activity monitor to estimate sleep duration. Participants on average lost 70 (±49) minutes of sleep (P = .0015) on nights following aerobic exercise and 27 (±78) minutes (P = .3) following resistance exercise relative to control nights. The odds ratio with confidence intervals of nocturnal hypoglycaemia occurring on nights following aerobic and resistance exercise was 5.4 (1.3, 27.2) and 7.0 (1.7, 37.3), respectively. Aerobic exercise can cause sleep loss in T1D possibly from increased hypoglycaemia.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Dissonias/etiologia , Exercício Físico , Hipoglicemia/etiologia , Treinamento Resistido/efeitos adversos , Corrida , Centros Médicos Acadêmicos , Actigrafia , Adulto , Glicemia/análise , Estudos de Coortes , Terapia Combinada/efeitos adversos , Estudos Cross-Over , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Dissonias/complicações , Humanos , Hipoglicemia/fisiopatologia , Hipoglicemia/prevenção & controle , Sistemas de Infusão de Insulina/efeitos adversos , Monitorização Ambulatorial , Consumo de Oxigênio , Projetos Piloto
16.
Surgery ; 159(1): 226-39, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26531237

RESUMO

BACKGROUND: The multidimensional nomogram calculating the upper limit of normal PTH (maxPTH) model identifies a personalized upper limit of normal parathyroid hormone (PTH) and successfully predicts classical primary hyperparathyroidism (PHP). We aimed to assess whether maxPTH can distinguish normocalcemic PHP (NCPHP) from secondary hyperparathyroidism (SHP), including subjects who underwent bariatric surgery (BrS). METHODS: A total of 172 subjects with 359 complete datasets of serum calcium (Ca), 25-OH vitamin D, and intact PTH from Oregon were analyzed: 123 subjects (212 datasets) with PHP and 47 (143) with SHP, including 28 (100) with previous BrS. An improved prediction model, MultIdimensional evaluation for Primary hyperparaTHyroidism (Mi-PTH), was created with the same variables as maxPTH by the use of a combined cohort (995 subjects) including participants from previous studies. RESULTS: In the Oregon cohort, maxPTH's sensitivity was 100% for classical PHP and 89% for NCPHP, but only 50% for normohormonal PHP (NHPHP) and 40% specific for SHP. In comparison, although sensitivity for NCPHP was similar (89%), Mi-PTH vastly improved SHP specificity (85%). In the combined cohort, Mi-PTH had better sensitivity of 98.5% (vs 95%) and specificity 97% (vs 85%). CONCLUSION: MaxPTH was sensitive in detecting PHP; however, there was low specificity for SHP, especially in patients who underwent BrS. The creation of Mi-PTH provided improved performance measures but requires further prospective evaluation.


Assuntos
Hiperparatireoidismo Primário/sangue , Hiperparatireoidismo Secundário/sangue , Nomogramas , Hormônio Paratireóideo/sangue , Adulto , Idoso , Cirurgia Bariátrica , Cálcio/sangue , Feminino , Humanos , Hiperparatireoidismo Primário/diagnóstico , Hiperparatireoidismo Secundário/diagnóstico , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Valor Preditivo dos Testes , Prognóstico , Valores de Referência
17.
Annu Int Conf IEEE Eng Med Biol Soc ; 2016: 2270-2273, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28324962

RESUMO

The Artificial Pancreas (AP) is a new technology for helping people with type 1 diabetes to better control their glucose levels through automated delivery of insulin and optionally glucagon in response to sensed glucose levels. In a dual hormone AP, insulin and glucagon are delivered automatically to the body based on glucose sensor measurements using a control algorithm that calculates the amount of hormones to be infused. A dual-hormone MPC may deliver insulin continuously; however, it must avoid continuous delivery of glucagon because nausea can occur from too much glucagon. In this paper, we propose a novel dual-hormone (DH) switching model predictive control and compare it with a single-hormone (SH) MPC. We extended both MPCs by integrating an exercise model and compared performance with and without the exercise model included. Results were obtained on a virtual patient population undergoing a simulated exercise event using a mathematical glucoregulatory model that includes exercise. Time spent in hypoglycemia is significantly less with the DH-MPC than the SH-MPC (p=0.0022). Additionally, including the exercise model in the DH-MPC can help prevent hypoglycemia (p <; 0.001).


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Hipoglicemiantes , Modelos Biológicos , Pâncreas Artificial , Glicemia , Glucagon , Humanos , Hipoglicemia/tratamento farmacológico , Insulina , Sistemas de Infusão de Insulina
18.
J Diabetes Sci Technol ; 9(6): 1175-84, 2015 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-26438720

RESUMO

In this article, we present several important contributions necessary for enabling an artificial endocrine pancreas (AP) system to better respond to exercise events. First, we show how exercise can be automatically detected using body-worn accelerometer and heart rate sensors. During a 22 hour overnight inpatient study, 13 subjects with type 1 diabetes wearing a Zephyr accelerometer and heart rate monitor underwent 45 minutes of mild aerobic treadmill exercise while controlling their glucose levels using sensor-augmented pump therapy. We used the accelerometer and heart rate as inputs into a validated regression model. Using this model, we were able to detect the exercise event with a sensitivity of 97.2% and a specificity of 99.5%. Second, from this same study, we show how patients' glucose declined during the exercise event and we present results from in silico modeling that demonstrate how including an exercise model in the glucoregulatory model improves the estimation of the drop in glucose during exercise. Last, we present an exercise dosing adjustment algorithm and describe parameter tuning and performance using an in silico glucoregulatory model during an exercise event.


Assuntos
Actigrafia/instrumentação , Algoritmos , Diabetes Mellitus Tipo 1/tratamento farmacológico , Cálculos da Dosagem de Medicamento , Teste de Esforço , Exercício Físico , Glucagon/administração & dosagem , Frequência Cardíaca , Hipoglicemiantes/administração & dosagem , Insulina/administração & dosagem , Pâncreas Artificial , Biomarcadores/sangue , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Simulação por Computador , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/fisiopatologia , Metabolismo Energético , Desenho de Equipamento , Glucagon/farmacocinética , Humanos , Hipoglicemiantes/farmacocinética , Insulina/farmacocinética , Modelos Lineares , Modelos Biológicos , Valor Preditivo dos Testes , Fatores de Tempo
19.
Diabetes Care ; 38(11): 2115-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26341131

RESUMO

OBJECTIVE: To evaluate subjects with type 1 diabetes for hepatic glycogen depletion after repeated doses of glucagon, simulating delivery in a bihormonal closed-loop system. RESEARCH DESIGN AND METHODS: Eleven adult subjects with type 1 diabetes participated. Subjects underwent estimation of hepatic glycogen using (13)C MRS. MRS was performed at the following four time points: fasting and after a meal at baseline, and fasting and after a meal after eight doses of subcutaneously administered glucagon at a dose of 2 µg/kg, for a total mean dose of 1,126 µg over 16 h. The primary and secondary end points were, respectively, estimated hepatic glycogen by MRS and incremental area under the glucose curve for a 90-min interval after glucagon administration. RESULTS: In the eight subjects with complete data sets, estimated glycogen stores were similar at baseline and after repeated glucagon doses. In the fasting state, glycogen averaged 21 ± 3 g/L before glucagon administration and 25 ± 4 g/L after glucagon administration (mean ± SEM) (P = NS). In the fed state, glycogen averaged 40 ± 2 g/L before glucagon administration and 34 ± 4 g/L after glucagon administration (P = NS). With the use of an insulin action model, the rise in glucose after the last dose of glucagon was comparable to the rise after the first dose, as measured by the 90-min incremental area under the glucose curve. CONCLUSIONS: In adult subjects with well-controlled type 1 diabetes (mean A1C 7.2%), glycogen stores and the hyperglycemic response to glucagon administration are maintained even after receiving multiple doses of glucagon. This finding supports the safety of repeated glucagon delivery in the setting of a bihormonal closed-loop system.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Glucagon/uso terapêutico , Hormônios/uso terapêutico , Hipoglicemia/terapia , Glicogênio Hepático/metabolismo , Adulto , Glicemia/análise , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/sangue , Retroalimentação Fisiológica , Feminino , Glucagon/administração & dosagem , Hormônios/administração & dosagem , Humanos , Hipoglicemia/prevenção & controle , Insulina/administração & dosagem , Insulina/uso terapêutico , Glicogênio Hepático/deficiência , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...