artificial pancreas
Recently Published Documents


TOTAL DOCUMENTS

1101
(FIVE YEARS 352)

H-INDEX

59
(FIVE YEARS 11)

Sensors ◽  
2022 ◽  
Vol 22 (2) ◽  
pp. 466
Author(s):  
John Daniels ◽  
Pau Herrero ◽  
Pantelis Georgiou

Current artificial pancreas (AP) systems are hybrid closed-loop systems that require manual meal announcements to manage postprandial glucose control effectively. This poses a cognitive burden and challenge to users with T1D since this relies on frequent user engagement to maintain tight glucose control. In order to move towards fully automated closed-loop glucose control, we propose an algorithm based on a deep learning framework that performs multitask quantile regression, for both meal detection and carbohydrate estimation. Our proposed method is evaluated in silico on 10 adult subjects from the UVa/Padova simulator with a Bio-inspired Artificial Pancreas (BiAP) control algorithm over a 2 month period. Three different configurations of the AP are evaluated -BiAP without meal announcement (BiAP-NMA), BiAP with meal announcement (BiAP-MA), and BiAP with meal detection (BiAP-MD). We present results showing an improvement of BiAP-MD over BiAP-NMA, demonstrating 144.5 ± 6.8 mg/dL mean blood glucose level (−4.4 mg/dL, p< 0.01) and 77.8 ± 6.3% mean time between 70 and 180 mg/dL (+3.9%, p< 0.001). This improvement in control is realised without a significant increase in mean in hypoglycaemia (+0.1%, p= 0.4). In terms of detection of meals and snacks, the proposed method on average achieves 93% precision and 76% recall with a detection delay time of 38 ± 15 min (92% precision, 92% recall, and 37 min detection time for meals only). Furthermore, BiAP-MD handles hypoglycaemia better than BiAP-MA based on CVGA assessment with fewer control errors (10% vs. 20%). This study suggests that multitask quantile regression can improve the capability of AP systems for postprandial glucose control without increasing hypoglycaemia.


2021 ◽  
Author(s):  
Rodrigo Vilanova ◽  
Anderson Jefferson Cerqueira

The number of children, adolescents, and adults living with diabetes increases annually due to the lack of physical activity, poor diet habits, stress, and genetic factors, and there are greater numbers in low-income countries. Therefore, the aim of this article is to present a proposal for a methodology for developing a pancreas using artificial intelligence to control the required doses of insulin for a patient with type 1 diabetes (T1D), according to data received from monitoring sensors. The information collected can be used by physicians to make medication changes and improve patients’ glucose control using insulin pumps for optimum performance. Therefore, using the model proposed in this work, the patient is offered a gain in glucose control and, therefore, an improvement in quality of life, as well as in the costs related to hospitalization.


Author(s):  
Ishita Rai ◽  
Anil Wanjari ◽  
Sourya Acharya

The discovery of insulin was 100 years old till 2021. Insulin, the first diabetic medication, is now the safest and most effective glucose-lowering medication available. Despite its efficacy, the most significant challenge with insulin has been the prevalence of hypoglycemia, which has resulted in the majority of patients being prescribed optimum dosages. Insulin delivery devices include syringes, pens, and pumps. Soon, artificial pancreas (AP) by using a very closed-loop delivery method will be a big step towards the advancement of insulin delivery devices. This article looks at the invention of syringes, disposable, long-lasting pens, and smart connected pens, continuous intraperitoneal insulin infusion (CIPII) and patch insulin pumps, artificial pancreas and other medical devices. Hence, insulin administration that is both minimally invasive and non-invasive towards the advancement is required. We review the available information on the evolution of insulin delivery systems, focusing on the advantages and disadvantages of technology as well as anticipated advances. Due to the wide variety of technological solutions accessible via the international platform, only the most common methods essential to the patient’s care are detailed here in the article.


2021 ◽  
pp. 193229682110591
Author(s):  
John P. Corbett ◽  
Jose Garcia-Tirado ◽  
Patricio Colmegna ◽  
Jenny L. Diaz Castaneda ◽  
Marc D. Breton

Introduction: Hyperglycemia following meals is a recurring challenge for people with type 1 diabetes, and even the most advanced available automated systems currently require manual input of carbohydrate amounts. To progress toward fully automated systems, we present a novel control system that can automatically deliver priming boluses and/or anticipate eating behaviors to improve postprandial full closed-loop control. Methods: A model predictive control (MPC) system was enhanced by an automated bolus system reacting to early glucose rise and/or a multistage MPC (MS-MPC) framework to anticipate historical patterns. Priming was achieved by detecting large glycemic disturbances, such as meals, and delivering a fraction of the patient’s total daily insulin (TDI) modulated by the disturbance’s likelihood (bolus priming system [BPS]). In the anticipatory module, glycemic disturbance profiles were generated from historical data using clustering to group days with similar behaviors; the probability of each cluster is then evaluated at every controller step and informs the MS-MPC framework to anticipate each profile. We tested four configurations: MPC, MPC + BPS, MS-MPC, and MS-MPC + BPS in simulation to contrast the effect of each controller module. Results: Postprandial time in range was highest for MS-MPC + BPS: 60.73 ± 25.39%, but improved with each module: MPC + BPS: 56.95±25.83 and MS-MPC: 54.83 ± 26.00%, compared with MPC: 51.79 ± 26.12%. Exposure to hypoglycemia was maintained for all controllers (time below 70 mg/dL <0.5%), and improvement came primarily from a reduction in postprandial time above range (MS-MPC + BPS: 39.10 ± 25.32%, MPC + BPS: 42.99 ± 25.81%, MS-MPC: 45.09 ± 25.96%, MPC: 48.18 ± 26.09%). Conclusions: The BPS and anticipatory disturbance profiles improved blood glucose control and were most efficient when combined.


2021 ◽  
pp. 193229682110591
Author(s):  
Xiaoyu Sun ◽  
Mudassir Rashid ◽  
Nicole Hobbs ◽  
Rachel Brandt ◽  
Mohammad Reza Askari ◽  
...  

Background: Adaptive model predictive control (MPC) algorithms that recursively update the glucose prediction model are shown to be promising in the development of fully automated multivariable artificial pancreas systems. However, the recursively updated glycemic prediction models do not explicitly consider prior knowledge in the identification of the model parameters. Prior information of the glycemic effects of meals and physical activity can improve model accuracy and yield better glycemic control algorithms. Methods: A glucose prediction model based on regularized partial least squares (rPLS) method where the prior information is encoded as the regularization term is developed to provide accurate predictions of the future glucose concentrations. An adaptive MPC is developed that incorporates dynamic trajectories for the glucose setpoint and insulin dosing constraints based on the estimated plasma insulin concentration (PIC). The proposed adaptive MPC algorithm is robust to disturbances caused by unannounced meals and physical activities even in cases with missing glucose measurements. The effectiveness of the proposed adaptive MPC based on rPLS is investigated with in silico subjects of the multivariable glucose-insulin-physiological variables simulator (mGIPsim). Results: The efficacy of the proposed adaptive MPC strategy in regulating the blood glucose concentration (BGC) of people with T1DM is assessed using the average percent time in range (TIR) for glucose, defined as 70 to 180 mg/dL inclusive, and the average percent time in hypoglycemia (<70 and >54 mg/dL) and level 2 hypoglycemia (≤54 mg/dL). The TIR for a cohort of 20 virtual subjects of mGIPsim is 81.9% ± 7.4% (with no hypoglycemia or severe hypoglycemia) for the proposed MPC compared with 73.9% ± 7.6% (0.2% ± 0.1% in hypoglycemia and 0.1% ± 0.1% in level 2 hypoglycemia) for an MPC based on a recursive autoregressive exogenous (ARX) model. Conclusions: The adaptive MPC algorithm that incorporates prior knowledge in the recursive updating of the glucose prediction model can contribute to the development of fully automated artificial pancreas systems that can mitigate meal and physical activity disturbances.


2021 ◽  
pp. 193229682110600
Author(s):  
Ryan Armiger ◽  
Monika Reddy ◽  
Nick S. Oliver ◽  
Pantelis Georgiou ◽  
Pau Herrero

Background: User-developed automated insulin delivery systems, also referred to as do-it-yourself artificial pancreas systems (DIY APS), are in use by people living with type 1 diabetes. In this work, we evaluate, in silico, the DIY APS Loop control algorithm and compare it head-to-head with the bio-inspired artificial pancreas (BiAP) controller for which clinical data are available. Methods: The Python version of the Loop control algorithm called PyLoopKit was employed for evaluation purposes. A Python-MATLAB interface was created to integrate PyLoopKit with the UVa-Padova simulator. Two configurations of BiAP (non-adaptive and adaptive) were evaluated. In addition, the Tandem Basal-IQ predictive low-glucose suspend was used as a baseline algorithm. Two scenarios with different levels of variability were used to challenge the algorithms on the adult (n = 10) and adolescent (n = 10) virtual cohorts of the simulator. Results: Both BiAP and Loop improve, or maintain, glycemic control when compared with Basal-IQ. Under the scenario with lower variability, BiAP and Loop perform relatively similarly. However, BiAP, and in particular its adaptive configuration, outperformed Loop in the scenario with higher variability by increasing the percentage time in glucose target range 70-180 mg/dL (BiAP-Adaptive vs Loop vs Basal-IQ) (adults: 89.9% ± 3.2%* vs 79.5% ± 5.3%* vs 67.9% ± 8.3%; adolescents: 74.6 ± 9.5%* vs 53.0% ± 7.7% vs 55.4% ± 12.0%, where * indicates the significance of P < .05 calculated in sequential order) while maintaining the percentage time below range (adults: 0.89% ± 0.37% vs 1.72% ± 1.26% vs 3.41 ± 1.92%; adolescents: 2.87% ± 2.77% vs 4.90% ± 1.92% vs 4.17% ± 2.74%). Conclusions: Both Loop and BiAP algorithms are safe and improve glycemic control when compared, in silico, with Basal-IQ. However, BiAP appears significantly more robust to real-world challenges by outperforming Loop and Basal-IQ in the more challenging scenario.


Sign in / Sign up

Export Citation Format

Share Document