Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
IEEE Trans Control Syst Technol ; 28(6): 2600-2607, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33762804

RESUMO

While artificial pancreas (AP) systems are expected to improve the quality of life among people with type 1 diabetes mellitus (T1DM), the design of convenient systems that optimize the user experience, especially for those with active lifestyles, such as children and adolescents, still remains an open research question. In this work, we introduce an embeddable design and implementation of model predictive control (MPC) of AP systems for people with T1DM that significantly reduces the weight and on-body footprint of the AP system. The embeddable controller is based on a zone MPC that has been evaluated in multiple clinical studies. The proposed embedded zone MPC features a simpler design of the periodic safe zone in the cost function and the utilization of state-of-the-art alternating minimization algorithms for solving the convex programming problems inherent to MPC with linear models subject to convex constraints. Off-line closed-loop data generated by the FDA-accepted UVA/Padova simulator is used to select an optimization algorithm and corresponding tuning parameters. Through hardware-in-the-loop in silico results on a limited-resource Arduino Zero (Feather M0) platform, we demonstrate the potential of the proposed embedded MPC. In spite of resource limitations, our embedded zone MPC manages to achieve comparable performance of that of the full-version zone MPC implemented in a 64-bit desktop for scenarios with/without meal-disturbance compensations. Metrics for performance comparison included median percent time in the euglycemic ([70, 180] mg/dL range) of 84.3% vs. 83.1% for announced meals, with an equivalence test yielding p = 0.0013 and 66.2% vs. 66.0% for unannounced meals with p = 0.0028.

2.
J Clin Endocrinol Metab ; 105(4)2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31714583

RESUMO

BACKGROUND: Postbariatric hypoglycemia (PBH) can threaten safety and reduce quality of life. Current therapies are incompletely effective. METHODS: Patients with PBH were enrolled in a double-blind, placebo-controlled, crossover trial to evaluate a closed-loop glucose-responsive automated glucagon delivery system designed to reduce severe hypoglycemia. A hypoglycemia detection and mitigation algorithm was embedded in the artificial pancreas system connected to a continuous glucose monitor (CGM, Dexcom) driving a patch infusion pump (Insulet) filled with liquid investigational glucagon (Xeris) or placebo (vehicle). Sensor/plasma glucose responses to mixed meal were assessed during 2 study visits. The system delivered up to 2 doses of study drug (300/150 µg glucagon or equal-volume vehicle) if triggered by the algorithm. Rescue dextrose was given for plasma glucose <55 mg/dL or neuroglycopenia. RESULTS: Twelve participants (11 females/1 male, age 52 ± 2, 8 ± 1 years postsurgery, mean ± SEM) completed all visits. Predictive hypoglycemia alerts prompted automated drug delivery postmeal, when sensor glucose was 114 ± 7 vs 121 ± 5 mg/dL (P = .39). Seven participants required rescue glucose after vehicle but not glucagon (P = .008). Five participants had severe hypoglycemia (<55 mg/dL) after vehicle but not glucagon (P = .03). Nadir plasma glucose was higher with glucagon vs vehicle (67 ± 3 vs 59 ± 2 mg/dL, P = .004). Plasma glucagon rose after glucagon delivery (1231 ± 187 vs 16 ± 1 pg/mL at 30 minutes, P = .001). No rebound hyperglycemia occurred. Transient infusion site discomfort was reported with both glucagon (n = 11/12) and vehicle (n = 10/12). No other adverse events were observed. CONCLUSION: A CGM-guided closed-loop rescue system can detect imminent hypoglycemia and deliver glucagon, reducing severe hypoglycemia in PBH. CLINICAL TRIALS REGISTRATION: NCT03255629.


Assuntos
Cirurgia Bariátrica/efeitos adversos , Fármacos Gastrointestinais/administração & dosagem , Glucagon/administração & dosagem , Hipoglicemia/tratamento farmacológico , Obesidade Mórbida/cirurgia , Algoritmos , Estudos Cross-Over , Método Duplo-Cego , Feminino , Seguimentos , Humanos , Hipoglicemia/etiologia , Hipoglicemia/patologia , Masculino , Pessoa de Meia-Idade , Prognóstico
3.
Diabetes Technol Ther ; 21(9): 485-492, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31225739

RESUMO

Background: Food choices are essential to successful glycemic control for people with diabetes. We compared the impact of three carbohydrate-rich meals on the postprandial glycemic response in adults with type 1 diabetes (T1D). Methods: We performed a randomized crossover study in 12 adults with T1D (age 58.7 ± 14.2 years, baseline hemoglobin A1c 7.5% ± 1.3%) comparing the postprandial glycemic response to three meals using continuous glucose monitoring: (1) "higher protein" pasta containing 10 g protein/serving, (2) regular pasta with 7 g protein/serving, and (3) extra-long grain white rice. All meals contained 42 g carbohydrate; were served with homemade tomato sauce, green salad, and balsamic dressing; and were repeated twice in random order. After their insulin bolus, subjects were observed in clinic for 5 h. Linear mixed effects models were used to assess the glycemic response. Results: Compared with white rice, peak glucose levels were significantly lower for higher protein pasta (-32.6 mg/dL; 95% CI -48.4 to -17.2; P < 0.001) and regular pasta (-43.2 mg/dL, 95% CI -58.7 to -27.7; P < 0.001). The difference between the two types of pastas did not reach statistical significance (-11 mg/dL; 95% CI -24.1 to 3.4; P = 0.17). Total glucose area under the curve was also significantly higher for white rice compared with both pastas (P < 0.001 for both comparisons). Conclusions: This exploratory study concluded that different food types of similar macronutrient content (e.g., rice and pasta) generate significantly different postprandial glycemic responses in persons with T1D. These results provide useful insights into the impact of food choices on and optimization of glucose control. Clinical Trial Registry: clinicaltrials.gov NCT03362151.


Assuntos
Glicemia/análise , Diabetes Mellitus Tipo 1/sangue , Grão Comestível/metabolismo , Oryza/metabolismo , Período Pós-Prandial/fisiologia , Adulto , Automonitorização da Glicemia , Estudos Cross-Over , Diabetes Mellitus Tipo 1/terapia , Carboidratos da Dieta/administração & dosagem , Feminino , Índice Glicêmico , Humanos , Hipoglicemiantes/administração & dosagem , Insulina/administração & dosagem , Masculino , Refeições , Pessoa de Meia-Idade
4.
Diabetes Technol Ther ; 21(1): 35-43, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30547670

RESUMO

BACKGROUND: There is an unmet need for a modular artificial pancreas (AP) system for clinical trials within the existing regulatory framework to further AP research projects from both academia and industry. We designed, developed, and tested the interoperable artificial pancreas system (iAPS) smartphone app that can interface wirelessly with leading continuous glucose monitors (CGM), insulin pump devices, and decision-making algorithms while running on an unlocked smartphone. METHODS: After algorithm verification, hazard and mitigation analysis, and complete system verification of iAPS, six adults with type 1 diabetes completed 1 week of sensor-augmented pump (SAP) use followed by 48 h of AP use with the iAPS, a Dexcom G5 CGM, and either a Tandem or Insulet insulin pump in an investigational device exemption study. The AP system was challenged by participants performing extensive walking without exercise announcement to the controller, multiple large meals eaten out at restaurants, two overnight periods, and multiple intentional connectivity interruptions. RESULTS: Even with these intentional challenges, comparison of the SAP phase with the AP study showed a trend toward improved time in target glucose range 70-180 mg/dL (78.8% vs. 83.1%; P = 0.31), and a statistically significant reduction in time below 70 mg/dL (6.1% vs. 2.2%; P = 0.03). The iAPS system performed reliably and showed robust connectivity with the peripheral devices (99.8% time connected to CGM and 94.3% time in closed loop) while requiring limited user intervention. CONCLUSIONS: The iAPS system was safe and effective in regulating glucose levels under challenging conditions and is suitable for use in unconstrained environments.


Assuntos
Automonitorização da Glicemia/métodos , Diabetes Mellitus Tipo 1/terapia , Sistemas de Infusão de Insulina , Aplicativos Móveis , Pâncreas Artificial , Adulto , Algoritmos , Glicemia/efeitos dos fármacos , Diabetes Mellitus Tipo 1/sangue , Feminino , Humanos , Hipoglicemiantes/administração & dosagem , Insulina/administração & dosagem , Masculino , Pesquisa , Smartphone , Resultado do Tratamento
5.
IEEE Trans Biomed Eng ; 65(3): 575-586, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28541890

RESUMO

OBJECTIVE: The development of artificial pancreas (AP) technology for deployment in low-energy, embedded devices is contingent upon selecting an efficient control algorithm for regulating glucose in people with type 1 diabetes mellitus. In this paper, we aim to lower the energy consumption of the AP by reducing controller updates, that is, the number of times the decision-making algorithm is invoked to compute an appropriate insulin dose. METHODS: Physiological insights into glucose management are leveraged to design an event-triggered model predictive controller (MPC) that operates efficiently, without compromising patient safety. The proposed event-triggered MPC is deployed on a wearable platform. Its robustness to latent hypoglycemia, model mismatch, and meal misinformation is tested, with and without meal announcement, on the full version of the US-FDA accepted UVA/Padova metabolic simulator. RESULTS: The event-based controller remains on for 18 h of 41 h in closed loop with unannounced meals, while maintaining glucose in 70-180 mg/dL for 25 h, compared to 27 h for a standard MPC controller. With meal announcement, the time in 70-180 mg/dL is almost identical, with the controller operating a mere 25.88% of the time in comparison with a standard MPC. CONCLUSION: A novel control architecture for AP systems enables safe glycemic regulation with reduced processor computations. SIGNIFICANCE: Our proposed framework integrated seamlessly with a wide variety of popular MPC variants reported in AP research, customizes tradeoff between glycemic regulation and efficacy according to prior design specifications, and eliminates judicious prior selection of controller sampling times.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Modelos Estatísticos , Pâncreas Artificial , Algoritmos , Automonitorização da Glicemia , Humanos , Hipoglicemia/tratamento farmacológico , Hipoglicemia/prevenção & controle , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/uso terapêutico
6.
J Diabetes Sci Technol ; 11(6): 1070-1079, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29032732

RESUMO

BACKGROUND: Continuous glucose monitoring (CGM) systems are increasingly becoming essential components in type 1 diabetes mellitus (T1DM) management. Current CGM technology requires frequent calibration to ensure accurate sensor performance. The accuracy of these systems is of great importance since medical decisions are made based on monitored glucose values and trends. METHODS: In this work, we introduce a calibration strategy that is augmented with a weekly updating feature. During the life cycle of the sensor, the calibration mechanism periodically estimates the parameters of a calibration model to fit self-monitoring blood glucose (SMBG) measurements. At the end of each week of use, an optimization problem that minimizes the sum of squared residuals between past reference and predicted blood glucose values is solved remotely to identify personalized calibration parameters. The newly identified parameters are used to initialize the calibration mechanism of the following week. RESULTS: The proposed method was evaluated using two sets of clinical data both consisting of 6 weeks of Dexcom G4 Platinum CGM data on 10 adults with T1DM (over 10 000 hours of CGM use), with seven SMBG data points per day measured by each subject in an unsupervised outpatient setting. Updating the calibration parameters using the history of calibration data indicated a positive trend of improving CGM performance. CONCLUSIONS: Although not statistically significant, the updating framework showed a relative improvement of CGM accuracy compared to the non-updating, static calibration method. The use of information collected for longer periods is expected to improve the performance of the sensor over time.


Assuntos
Algoritmos , Automonitorização da Glicemia/métodos , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/diagnóstico , Processamento de Sinais Assistido por Computador , Adulto , Biomarcadores/sangue , Glicemia/efeitos dos fármacos , Automonitorização da Glicemia/instrumentação , Automonitorização da Glicemia/normas , Calibragem , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/tratamento farmacológico , Desenho de Equipamento , Feminino , Humanos , Hipoglicemiantes/uso terapêutico , Análise dos Mínimos Quadrados , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Reprodutibilidade dos Testes , Fatores de Tempo , Transdutores
7.
IEEE Trans Biomed Eng ; 62(10): 2369-78, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25935026

RESUMO

This study presents a general closed-loop control strategy for optimal insulin delivery in type 1 Diabetes Mellitus (T1DM). The proposed control strategy aims toward an individualized optimal insulin delivery that consists of a patient-specific model predictive controller, a state estimator, a personalized scheduling level, and an open-loop optimization problem subjected to patient-specific process model and constraints. This control strategy can be also modified to address the case of limited patient data availability resulting in an "approximation" control strategy. Both strategies are validated in silico in the presence of predefined and unknown meal disturbances using both a novel mathematical model of glucose-insulin interactions and the UVa/Padova Simulator model as a virtual patient. The robustness of the control performance is evaluated under several conditions such as skipped meals, variability in the meal time, and metabolic uncertainty. The simulation results of the closed-loop validation studies indicate that the proposed control strategies can potentially achieve improved glycaemic control.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Insulina/administração & dosagem , Modelos Biológicos , Modelos Estatísticos , Pâncreas Artificial , Processamento de Sinais Assistido por Computador , Glicemia/análise , Simulação por Computador , Humanos , Insulina/uso terapêutico , Sistemas de Infusão de Insulina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA