1
|
Romeres D, Ruchi FNU, Breton MD, Basu A, DeBoer MD. Postprandial Glucose Turnover Differs Between Adolescents and Adults With Type 1 Diabetes: Triple Tracer Mixed Meal Study. J Clin Endocrinol Metab 2025; 110:730-738. [PMID: 39169876 DOI: 10.1210/clinem/dgae585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/08/2024] [Accepted: 08/20/2024] [Indexed: 08/23/2024]
Abstract
CONTEXT Insulin sensitivity (SI) varies with age in type 1 diabetes (T1D). OBJECTIVE To compare postprandial glucose turnover and SI between adolescents and adults with T1D. DESIGN This cross-sectional comparison at a clinical research unit included 21 early adolescents with T1D (T1D-adol) (12 F; age, 11.5 ± 0.5 years; BMI 19 ± 2 kg/m2), 13 adults with T1D (T1D-adult) (5 F; 37.8 ± 9.1 years; BMI 27 ± 2 kg/m2), and 14 anthropometrically matched adults without diabetes (ND) (7 F; 26.9 ± 7.0 years; BMI 25 ± 2.5 kg/m2). Using triple tracer mixed meal and oral glucose models, SI in T1D-adol and T1D-adult was compared. RESULTS Postprandial glucose excursions were not different in T1D-adol vs T1D-adult (P = .111) but higher than in ND (P < .01). Insulin excursions were also similar in T1D-adol vs T1D-adult (P = .600) and they were both lower (P < .05) compared to ND, while glucagon excursions were lower (P < .01) in T1D-adol than in T1D-adult and ND. Integrated rates of endogenous glucose production and glucose disappearance were lower in T1D-adol than in T1D-adult and in ND vs T1D-adult but did not differ between T1D-adol and ND. Meal glucose appearance did not differ between groups. While SI in T1D-adol vs ND was similar (P = .299), it was higher in T1D-adol and ND vs T1D-adult (P < .01). CONCLUSION We report differences in parameters of postprandial glucose turnover and insulin sensitivity between adults and early adolescents with T1D that could, at least in part, be due to the shorter duration of diabetes among T1D-adol. These data support the concept that over time with T1D, endogenous glucose production increases and SI deteriorates.
Collapse
Affiliation(s)
- Davide Romeres
- Department of Medicine, Division of Endocrinology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - F N U Ruchi
- Department of Medicine, Division of Endocrinology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Marc D Breton
- Center for Diabetes Technology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Ananda Basu
- Department of Medicine, Division of Endocrinology, University of Alabama-Birmingham Birmingham, AL 35233, USA
| | - Mark D DeBoer
- Center for Diabetes Technology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| |
Collapse
|
2
|
Mohan S, Christie HE, Laurenti MC, Egan AM, Bailey KR, Cobelli C, Dalla Man C, Vella A. Abnormal glucagon secretion contributes to a longitudinal decline in glucose tolerance. J Clin Endocrinol Metab 2025:dgae915. [PMID: 39758010 DOI: 10.1210/clinem/dgae915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
CONTEXT Defects in insulin secretion and action contribute to the progression of prediabetes to diabetes. However, the contribution of α-cell dysfunction to this process has been unclear. OBJECTIVE Understand the relative contributions of α-cell and β-cell dysfunction to declining glucose tolerance. DESIGN Longitudinal, community-based observational study. SETTING Clinical Research Unit at an Academic Medical Center. PATIENTS/PARTICIPANTS We studied 96 subjects without diabetes (55 ± 1 years; BMI 27.7 ± 0.4 Kg/M2) on 2 occasions, 3 years apart using an oral 75g glucose challenge. Indices for insulin secretion and action were estimated using the oral minimal model. Glucagon secretion rate (GSR) was estimated by deconvolution from peripheral glucagon concentrations. INTERVENTION This was an observational study. MAIN OUTCOME MEASURE Glucose tolerance status (categorical variable) and then symmetrical percent change in peak and 120-minute glucose (post-OGTT) concentrations (continuous variables). RESULTS 32 subjects progressed from normal to Impaired Glucose Tolerance (IGT) or from IGT to type 2 diabetes. Disposition Index (DI) declined in the progressors (568±98 vs. 403±65 10-4 dl/kg/min per μU/ml, baseline vs. 3-years p=0.04). α-cell suppression by glucose (δGSR/δglucose) did not change in the non-progressors (1.5±0.1 vs. 1.3±0.1 nmol/min/L, p=0.37) but decreased (1.0±0.2 vs. 0.8±0.2 nmol/min/L, p<0.01) in those who progressed. Analysis of the entire cohort showed that DI and δGSR/δglucose were independently and inversely correlated with an increase in glycemic excursion. CONCLUSIONS These data show that α-cell dysfunction accompanies a decline in β-cell function as IGT or overt type 2 diabetes develops.
Collapse
Affiliation(s)
- Sneha Mohan
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Hannah E Christie
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Marcello C Laurenti
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Aoife M Egan
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Kent R Bailey
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Claudio Cobelli
- Department of Women and Children's Health, University of Padova, Padova, Italy
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
3
|
Farman M, Hasan A, Xu C, Nisar KS, Hincal E. Computational techniques to monitoring fractional order type-1 diabetes mellitus model for feedback design of artificial pancreas. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108420. [PMID: 39303363 DOI: 10.1016/j.cmpb.2024.108420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND AND OBJECTIVES In this paper, we developed a significant class of control issues regulated by nonlinear fractal order systems with input and output signals, our goal is to design a direct transcription method with impulsive instant order. Recent advances in the artificial pancreas system provide an emerging treatment option for type 1 diabetes. The performance of the blood glucose regulation directly relies on the accuracy of the glucose-insulin modeling. This work leads to the monitoring and assessment of comprehensive type-1 diabetes mellitus for controller design of artificial panaceas for the precision of the glucose-insulin glucagon in finite time with Caputo fractional approach for three primary subsystems. METHODS For the proposed model, we admire the qualitative analysis with equilibrium points lying in the feasible region. Model satisfied the biological feasibility with the Lipschitz criteria and linear growth is examined, considering positive solutions, boundedness and uniqueness at equilibrium points with Leray-Schauder results under time scale ideas. Within each subsystem, the virtual control input laws are derived by the application of input to state theorems and Ulam Hyers Rassias. RESULTS Chaotic Relation of Glucose insulin glucagon compartmental in the feasible region and stable in finite time interval monitoring is derived through simulations that are stable and bounded in the feasible regions. Additionally, as blood glucose is the only measurable state variable, the unscented power-law kernel estimator appropriately takes into account the significant problem of estimating inaccessible state variables that are bound to significant values for the glucose-insulin system. The comparative results on the simulated patients suggest that the suggested controller strategy performs remarkably better than the compared methods. CONCLUSION In the model under investigation, parametric uncertainties are identified since the glucose, insulin, and glucagon system's parameters are accurately measured numerically at different fractional order values. In terms of algorithm resilience and Caputo tracking in the presence of glucagon and insulin intake disturbance to maintain the glucose level. A comprehensive analysis of numerous difficult test issues is conducted in order to offer a thorough justification of the planned strategy to control the type 1 diabetes mellitus with designed the artificial pancreas.
Collapse
Affiliation(s)
- Muhammad Farman
- Department of Mathematics,Faculty of Science and Technology, Universitas Airlangga, Surabaya, Indonesia; Department of Computer Science and Mathematics, Lebanese American University, 1107-2020, Beirut, Lebanon
| | - Ali Hasan
- Department of Mathematics and Statistics, The University of Lahore, Pakistan
| | - Changjin Xu
- Guizhou Key Laboratory of Economics System Simulation, Guizhou University of Finance and Economics, Guiyang 550025, PR China.
| | - Kottakkaran Sooppy Nisar
- Department of Mathematics, College of Science and Humanities in Alkharj, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia; Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Jordan
| | - Evren Hincal
- Faculty of Arts and sciences, Department of Mathematics, Near East University, 99138 Nicosia, Cyprus
| |
Collapse
|
4
|
Villa-Tamayo MF, Colmegna P, Breton M. Validation of the UVA Simulation Replay Methodology Using Clinical Data: Reproducing a Randomized Clinical Trial. Diabetes Technol Ther 2024; 26:720-727. [PMID: 38662426 DOI: 10.1089/dia.2023.0595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Background: Computer simulators of human metabolism are powerful tools to design and validate new diabetes treatments. However, these platforms are often limited in the diversity of behaviors and glycemic conditions they can reproduce. Replay methodologies leverage field-collected data to create ad hoc simulation environments representative of real-life conditions. After formal validations of our method in prior publications, we demonstrate its capacity to reproduce a recent clinical trial. Methods: Using the replay methodology, an ensemble of replay simulators was generated using data from a randomized crossover clinical trial comparing the hybrid closed loop (HCL) and fully closed loop (FCL) control modalities in automated insulin delivery (AID), creating 64 subject/modality pairs. Each virtual subject was exposed to the alternate AID modality to compare the simulated versus observed glycemic outcomes. Equivalence tests were performed for time in, below, and above range (TIR, TBR, and TAR) and high and low blood glucose indices (HBGI and LBGI) considering equivalence margins corresponding to clinical significance. Results: TIR, TAR, LBGI, and HBGI showed statistical and clinical equivalence between the original and the simulated data; TBR failed the equivalence test. For example, in the HCL mode, simulated TIR was 84.89% versus an observed 84.31% (P = 0.0170, confidence interval [CI] [-3.96, 2.79]), and for FCL mode, TIR was 76.58% versus 77.41% (P = 0.0222, CI [-2.54, 4.20]). Conclusion: Clinical trial data confirm the prior in silico validation of the UVA replay method in predicting the glycemic impact of modified insulin treatments. This in vivo demonstration justifies the application of the replay method to the personalization and adaptation of treatment strategies in people with type 1 diabetes.
Collapse
Affiliation(s)
- María F Villa-Tamayo
- Center for Diabetes Technology, University of Virginia, Charlottesville, Virginia, USA
| | - Patricio Colmegna
- Center for Diabetes Technology, University of Virginia, Charlottesville, Virginia, USA
| | - Marc Breton
- Center for Diabetes Technology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
5
|
Sun T, Liu J, Chen CJ. Calibration algorithms for continuous glucose monitoring systems based on interstitial fluid sensing. Biosens Bioelectron 2024; 260:116450. [PMID: 38843770 DOI: 10.1016/j.bios.2024.116450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
Continuous glucose monitoring (CGM) is of great importance to the treatment and prevention of diabetes. As a proven commercial technology, electrochemical glucose sensor based on interstitial fluid (ISF) sensing has high sensitivity and wide detection range. Therefore, it has good promotion prospects in noninvasive or minimally-invasive CGM system. However, since there are concentration differences and time lag between glucose in plasma and ISF, the accuracy of this type of sensors are still limited. Typical calibration algorithms rely on simple linear regression which do not account for the variability of the sensitivity of sensors. To enhance the accuracy and stability of CGM based on ISF, optimization of calibration algorithm for sensors is indispensable. While there have been considerable researches on improving calibration algorithms for CGM, they have still received less attention. This article reviews the problem of typical calibration and presents the outstanding calibration algorithms in recent years. Finally, combined with existing research and emerging sensing technologies, this paper makes an outlook on the future calibration algorithms for CGM sensors.
Collapse
Affiliation(s)
- Tianyi Sun
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China.
| | - Jentsai Liu
- Research Center for Materials Science and Opti-Electronic Technology, College of Materials Science and Opti-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China.
| | - Ching Jung Chen
- 3 Research Center for Materials Science and Opti-Electronic Technology, School of Optoelectronics, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
6
|
Vella M, Mohan S, Christie H, Bailey KR, Cobelli C, Dalla Man C, Matveyenko A, Egan AM, Vella A. Diabetes-associated Genetic Variation in MTNR1B and Its Effect on Islet Function. J Endocr Soc 2024; 8:bvae130. [PMID: 39011323 PMCID: PMC11249077 DOI: 10.1210/jendso/bvae130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Indexed: 07/17/2024] Open
Abstract
Context Multiple common genetic variants have been associated with type 2 diabetes, but the mechanism by which they predispose to diabetes is incompletely understood. One such example is variation in MTNR1B, which implicates melatonin and its receptor in the pathogenesis of type 2 diabetes. Objective To characterize the effect of diabetes-associated genetic variation at rs10830963 in the MTNR1B locus on islet function in people without type 2 diabetes. Design The association of genetic variation at rs10830963 with glucose, insulin, C-peptide, glucagon, and indices of insulin secretion and action were tested in a cohort of 294 individuals who had previously undergone an oral glucose tolerance test (OGTT). Insulin sensitivity, β-cell responsivity to glucose, and Disposition Indices were measured using the oral minimal model. Setting The Clinical Research and Translation Unit at Mayo Clinic, Rochester, MN. Participants Two cohorts were utilized for this analysis: 1 cohort was recruited on the basis of prior participation in a population-based study in Olmsted County. The other cohort was recruited on the basis of TCF7L2 genotype at rs7903146 from the Mayo Biobank. Intervention Two-hour, 7-sample OGTT. Main Outcome Measures Fasting, nadir, and integrated glucagon concentrations. Results One or 2 copies of the G-allele at rs10830963 were associated with increased postchallenge glucose and glucagon concentrations compared to subjects with the CC genotype. Conclusion The effects of rs10830963 on glucose homeostasis and predisposition to type 2 diabetes are likely to be partially mediated through changes in α-cell function.
Collapse
Affiliation(s)
- Max Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Sneha Mohan
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Hannah Christie
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Kent R Bailey
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Claudio Cobelli
- Department of Women and Children's Health, University of Padova, 35128 Padova, Italy
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, 35128 Padova, Italy
| | - Aleksey Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Aoife M Egan
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
7
|
Kurt I, Krauhausen I, Spolaor S, van de Burgt Y. Predicting Blood Glucose Levels with Organic Neuromorphic Micro-Networks. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308261. [PMID: 38682442 PMCID: PMC11251550 DOI: 10.1002/advs.202308261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/05/2024] [Indexed: 05/01/2024]
Abstract
Accurate glucose prediction is vital for diabetes management. Artificial intelligence and artificial neural networks (ANNs) are showing promising results for reliable glucose predictions, offering timely warnings for glucose fluctuations. The translation of these software-based ANNs into dedicated computing hardware opens a route toward automated insulin delivery systems ultimately enhancing the quality of life for diabetic patients. ANNs are transforming this field, potentially leading to implantable smart prediction devices and ultimately to a fully artificial pancreas. However, this transition presents several challenges, including the need for specialized, compact, lightweight, and low-power hardware. Organic polymer-based electronics are a promising solution as they have the ability to implement the behavior of neural networks, operate at low voltage, and possess key attributes like flexibility, stretchability, and biocompatibility. Here, the study focuses on implementing software-based neural networks for glucose prediction into hardware systems. How to minimize network requirements, downscale the architecture, and integrate the neural network with electrochemical neuromorphic organic devices, meeting the strict demands of smart implants for in-body computation of glucose prediction is investigated.
Collapse
Affiliation(s)
- Ibrahim Kurt
- MicrosystemsInstitute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5612 AEThe Netherlands
| | - Imke Krauhausen
- MicrosystemsInstitute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5612 AEThe Netherlands
- Max Planck Institute for Polymer Research55128MainzGermany
| | - Simone Spolaor
- MicrosystemsInstitute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5612 AEThe Netherlands
| | - Yoeri van de Burgt
- MicrosystemsInstitute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5612 AEThe Netherlands
| |
Collapse
|
8
|
Welch AA, Farahani RA, Egan AM, Laurenti MC, Zeini M, Vella M, Bailey KR, Cobelli C, Dalla Man C, Matveyenko A, Vella A. Glucagon-like peptide-1 receptor blockade impairs islet secretion and glucose metabolism in humans. J Clin Invest 2023; 133:e173495. [PMID: 37751301 PMCID: PMC10645389 DOI: 10.1172/jci173495] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/12/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUNDProglucagon can be processed to glucagon-like peptide1 (GLP-1) within the islet, but its contribution to islet function in humans remains unknown. We sought to understand whether pancreatic GLP-1 alters islet function in humans and whether this is affected by type 2 diabetes.METHODSWe therefore studied individuals with and without type 2 diabetes on two occasions in random order. On one occasion, exendin 9-39, a competitive antagonist of the GLP-1 Receptor (GLP1R), was infused, while on the other, saline was infused. The tracer dilution technique ([3-3H] glucose) was used to measure glucose turnover during fasting and during a hyperglycemic clamp.RESULTSExendin 9-39 increased fasting glucose concentrations; fasting islet hormone concentrations were unchanged, but inappropriate for the higher fasting glucose observed. In people with type 2 diabetes, fasting glucagon concentrations were markedly elevated and persisted despite hyperglycemia. This impaired suppression of endogenous glucose production by hyperglycemia.CONCLUSIONThese data show that GLP1R blockade impairs islet function, implying that intra-islet GLP1R activation alters islet responses to glucose and does so to a greater degree in people with type 2 diabetes.TRIAL REGISTRATIONThis study was registered at ClinicalTrials.gov NCT04466618.FUNDINGThe study was primarily funded by NIH NIDDK DK126206. AV is supported by DK78646, DK116231 and DK126206. CDM was supported by MIUR (Italian Minister for Education) under the initiative "Departments of Excellence" (Law 232/2016).
Collapse
Affiliation(s)
- Andrew A. Welch
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Rahele A. Farahani
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Aoife M. Egan
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Marcello C. Laurenti
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Maya Zeini
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Max Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Kent R. Bailey
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Aleksey Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Cobelli C, Kovatchev B. Developing the UVA/Padova Type 1 Diabetes Simulator: Modeling, Validation, Refinements, and Utility. J Diabetes Sci Technol 2023; 17:1493-1505. [PMID: 37743740 PMCID: PMC10658679 DOI: 10.1177/19322968231195081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Arguably, diabetes mellitus is one of the best quantified human conditions. In the past 50 years, the metabolic monitoring technologies progressed from occasional assessment of average glycemia via HbA1c, through episodic blood glucose readings, to continuous glucose monitoring (CGM) producing data points every few minutes. The high-temporal resolution of CGM data enabled increasingly intensive treatments, from decision support assisting insulin injection or oral medication, to automated closed-loop control, known as the "artificial pancreas." Throughout this progress, mathematical models and computer simulation of the human metabolic system became indispensable for the technological progress of diabetes treatment, enabling every step, from assessment of insulin sensitivity via the now classic Minimal Model of Glucose Kinetics, to in silico trials replacing animal experiments, to automated insulin delivery algorithms. In this review, we follow these developments, beginning with the Minimal Model, which evolved through the years to become large and comprehensive and trigger a paradigm change in the design of diabetes optimization strategies: in 2007, we introduced a sophisticated model of glucose-insulin dynamics and a computer simulator equipped with a "population" of N = 300 in silico "subjects" with type 1 diabetes. In January 2008, in an unprecedented decision, the Food and Drug Administration (FDA) accepted this simulator as a substitute to animal trials for the pre-clinical testing of insulin treatment strategies. This opened the field for rapid and cost-effective development and pre-clinical testing of new treatment approaches, which continues today. Meanwhile, animal experiments for the purpose of designing new insulin treatment algorithms have been abandoned.
Collapse
Affiliation(s)
| | - Boris Kovatchev
- Center for Diabetes Technology,
University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
10
|
Moscoso-Vasquez M, Fabris C, Breton MD. Performance Effect of Adjusting Insulin Sensitivity for Model-Based Automated Insulin Delivery Systems. J Diabetes Sci Technol 2023; 17:1470-1481. [PMID: 37864340 PMCID: PMC10658700 DOI: 10.1177/19322968231206798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2023]
Abstract
BACKGROUND Model predictive control (MPC) has become one of the most popular control strategies for automated insulin delivery (AID) in type 1 diabetes (T1D). These algorithms rely on a prediction model to determine the best insulin dosing every sampling time. Although these algorithms have been shown to be safe and effective for glucose management through clinical trials, managing the ever-fluctuating relationship between insulin delivery and resulting glucose uptake (aka insulin sensitivity, IS) remains a challenge. We aim to evaluate the effect of informing an AID system with IS on the performance of the system. METHOD The University of Virginia (UVA) MPC control-based hybrid closed-loop (HCL) and fully closed-loop (FCL) system was used. One-day simulations at varying levels of IS were run with the UVA/Padova T1D Simulator. The AID system was informed with an estimated value of IS obtained through a mixed meal glucose tolerance test. Relevant controller parameters are updated to inform insulin dosing of IS. Performance of the HCL/FCL system with and without information of the changing IS was assessed using a novel performance metric penalizing the time outside the target glucose range. RESULTS Feedback in AID systems provides a certain degree tolerance to changes in IS. However, IS-informed bolus and basal dosing improve glycemic outcomes, providing increased protection against hyperglycemia and hypoglycemia according to the individual's physiological state. CONCLUSIONS The proof-of-concept analysis presented here shows the potentially beneficial effects on system performance of informing the AID system with accurate estimates of IS. In particular, when considering reduced IS, the informed controller provides increased protection against hyperglycemia compared with the naïve controller. Similarly, reduced hypoglycemia is obtained for situations with increased IS. Further tailoring of the adaptation schemes proposed in this work is needed to overcome the increased hypoglycemia observed in the more resistant cases and to optimize the performance of the adaptation method.
Collapse
Affiliation(s)
| | - Chiara Fabris
- Center for Diabetes Technology,
University of Virginia, Charlottesville, VA, USA
| | - Marc D. Breton
- Center for Diabetes Technology,
University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
11
|
Skantze V, Hjorth T, Wallman M, Brunius C, Dicksved J, Pelve EA, Esberg A, Vitale M, Giacco R, Costabile G, Bergia RE, Jirstrand M, Campbell WW, Riccardi G, Landberg R. Differential Responders to a Mixed Meal Tolerance Test Associated with Type 2 Diabetes Risk Factors and Gut Microbiota-Data from the MEDGI-Carb Randomized Controlled Trial. Nutrients 2023; 15:4369. [PMID: 37892445 PMCID: PMC10609681 DOI: 10.3390/nu15204369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/04/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
The global prevalence of type 2 diabetes mellitus (T2DM) has surged in recent decades, and the identification of differential glycemic responders can aid tailored treatment for the prevention of prediabetes and T2DM. A mixed meal tolerance test (MMTT) based on regular foods offers the potential to uncover differential responders in dynamical postprandial events. We aimed to fit a simple mathematical model on dynamic postprandial glucose data from repeated MMTTs among participants with elevated T2DM risk to identify response clusters and investigate their association with T2DM risk factors and gut microbiota. Data were used from a 12-week multi-center dietary intervention trial involving high-risk T2DM adults, comparing high- versus low-glycemic index foods within a Mediterranean diet context (MEDGICarb). Model-based analysis of MMTTs from 155 participants (81 females and 74 males) revealed two distinct plasma glucose response clusters that were associated with baseline gut microbiota. Cluster A, inversely associated with HbA1c and waist circumference and directly with insulin sensitivity, exhibited a contrasting profile to cluster B. Findings imply that a standardized breakfast MMTT using regular foods could effectively distinguish non-diabetic individuals at varying risk levels for T2DM using a simple mechanistic model.
Collapse
Affiliation(s)
- Viktor Skantze
- Fraunhofer-Chalmers Research Centre for Industrial Mathematics, 412 88 Gothenburg, Sweden (M.J.)
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, 412 96 Gothenburg, Sweden; (T.H.); (R.L.)
| | - Therese Hjorth
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, 412 96 Gothenburg, Sweden; (T.H.); (R.L.)
| | - Mikael Wallman
- Fraunhofer-Chalmers Research Centre for Industrial Mathematics, 412 88 Gothenburg, Sweden (M.J.)
| | - Carl Brunius
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, 412 96 Gothenburg, Sweden; (T.H.); (R.L.)
| | - Johan Dicksved
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, 750 07 Uppsala, Sweden
| | - Erik A. Pelve
- Department of Animal Nutrition and Management, Swedish University of Agricultural Sciences, 750 07 Uppsala, Sweden;
| | - Anders Esberg
- Department of Odontology, Umeå University, 901 87 Umeå, Sweden
| | - Marilena Vitale
- Diabetes, Nutrition and Metabolism Unit, Department of Clinical Medicine and Surgery, Federico II University, 80138 Naples, Italy; (M.V.); (R.G.); (G.C.); (G.R.)
| | - Rosalba Giacco
- Diabetes, Nutrition and Metabolism Unit, Department of Clinical Medicine and Surgery, Federico II University, 80138 Naples, Italy; (M.V.); (R.G.); (G.C.); (G.R.)
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy
| | - Giuseppina Costabile
- Diabetes, Nutrition and Metabolism Unit, Department of Clinical Medicine and Surgery, Federico II University, 80138 Naples, Italy; (M.V.); (R.G.); (G.C.); (G.R.)
| | - Robert E. Bergia
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA (W.W.C.)
| | - Mats Jirstrand
- Fraunhofer-Chalmers Research Centre for Industrial Mathematics, 412 88 Gothenburg, Sweden (M.J.)
| | - Wayne W. Campbell
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA (W.W.C.)
| | - Gabriele Riccardi
- Diabetes, Nutrition and Metabolism Unit, Department of Clinical Medicine and Surgery, Federico II University, 80138 Naples, Italy; (M.V.); (R.G.); (G.C.); (G.R.)
| | - Rikard Landberg
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, 412 96 Gothenburg, Sweden; (T.H.); (R.L.)
| |
Collapse
|
12
|
Prendin F, Pavan J, Cappon G, Del Favero S, Sparacino G, Facchinetti A. The importance of interpreting machine learning models for blood glucose prediction in diabetes: an analysis using SHAP. Sci Rep 2023; 13:16865. [PMID: 37803177 PMCID: PMC10558434 DOI: 10.1038/s41598-023-44155-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023] Open
Abstract
Machine learning has become a popular tool for learning models of complex dynamics from biomedical data. In Type 1 Diabetes (T1D) management, these models are increasingly been integrated in decision support systems (DSS) to forecast glucose levels and provide preventive therapeutic suggestions, like corrective insulin boluses (CIB), accordingly. Typically, models are chosen based on their prediction accuracy. However, since patient safety is a concern in this application, the algorithm should also be physiologically sound and its outcome should be explainable. This paper aims to discuss the importance of using tools to interpret the output of black-box models in T1D management by presenting a case-of-study on the selection of the best prediction algorithm to integrate in a DSS for CIB suggestion. By retrospectively "replaying" real patient data, we show that two long-short term memory neural networks (LSTM) (named p-LSTM and np-LSTM) with similar prediction accuracy could lead to different therapeutic decisions. An analysis with SHAP-a tool for explaining black-box models' output-unambiguously shows that only p-LSTM learnt the physiological relationship between inputs and glucose prediction, and should therefore be preferred. This is verified by showing that, when embedded in the DSS, only p-LSTM can improve patients' glycemic control.
Collapse
Affiliation(s)
- Francesco Prendin
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Jacopo Pavan
- Department of Information Engineering, University of Padova, Padova, Italy
- Department of Psychiatry and Neurobehavioral Sciences, Center for Diabetes Technology, University of Virginia, Charlottesville, VA, USA
| | - Giacomo Cappon
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Simone Del Favero
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Giovanni Sparacino
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Andrea Facchinetti
- Department of Information Engineering, University of Padova, Padova, Italy.
| |
Collapse
|
13
|
Albers D, Sirlanci M, Levine M, Claassen J, Nigoghossian CD, Hripcsak G. Interpretable physiological forecasting in the ICU using constrained data assimilation and electronic health record data. J Biomed Inform 2023; 145:104477. [PMID: 37604272 DOI: 10.1016/j.jbi.2023.104477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 08/08/2023] [Accepted: 08/18/2023] [Indexed: 08/23/2023]
Abstract
OBJECTIVE Prediction of physiological mechanics are important in medical practice because interventions are guided by predicted impacts of interventions. But prediction is difficult in medicine because medicine is complex and difficult to understand from data alone, and the data are sparse relative to the complexity of the generating processes. Computational methods can increase prediction accuracy, but prediction with clinical data is difficult because the data are sparse, noisy and nonstationary. This paper focuses on predicting physiological processes given sparse, non-stationary, electronic health record data in the intensive care unit using data assimilation (DA), a broad collection of methods that pair mechanistic models with inference methods. METHODS A methodological pipeline embedding a glucose-insulin model into a new DA framework, the constrained ensemble Kalman filter (CEnKF) to forecast blood glucose was developed. The data include tube-fed patients whose nutrition, blood glucose, administered insulins and medications were extracted by hand due to their complexity and to ensure accuracy. The model was estimated using an individual's data as if they arrived in real-time, and the estimated model was run forward producing a forecast. Both constrained and unconstrained ensemble Kalman filters were estimated to compare the impact of constraints. Constraint boundaries, model parameter sets estimated, and data used to estimate the models were varied to investigate their influence on forecasting accuracy. Forecasting accuracy was evaluated according to mean squared error between the model-forecasted glucose and the measurements and by comparing distributions of measured glucose and forecast ensemble means. RESULTS The novel CEnKF produced substantial gains in robustness and accuracy while minimizing the data requirements compared to the unconstrained ensemble Kalman filters. Administered insulin and tube-nutrition were important for accurate forecasting, but including glucose in IV medication delivery did not increase forecast accuracy. Model flexibility, controlled by constraint boundaries and estimated parameters, did influence forecasting accuracy. CONCLUSION Accurate and robust physiological forecasting with sparse clinical data is possible with DA. Introducing constrained inference, particularly on unmeasured states and parameters, reduced forecast error and data requirements. The results are not particularly sensitive to model flexibility such as constraint boundaries, but over or under constraining increased forecasting errors.
Collapse
Affiliation(s)
- David Albers
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, 80045, CO, USA; Department of Biomedical Engineering, University of Colorado Anschutz Medical Campus, Aurora, 80045, CO, USA; Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, 80045, CO, USA; Department of Biomedical Informatics, Columbia University, New York, 10032, NY, USA.
| | - Melike Sirlanci
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, 80045, CO, USA
| | - Matthew Levine
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, 91125, CA, USA
| | - Jan Claassen
- Division of Critical Care Neurology, Department of Neurology, Columbia University, New York, 10032, NY, USA
| | | | - George Hripcsak
- Department of Biomedical Informatics, Columbia University, New York, 10032, NY, USA
| |
Collapse
|
14
|
Phillips NE, Collet TH, Naef F. Uncovering personalized glucose responses and circadian rhythms from multiple wearable biosensors with Bayesian dynamical modeling. CELL REPORTS METHODS 2023; 3:100545. [PMID: 37671030 PMCID: PMC10475794 DOI: 10.1016/j.crmeth.2023.100545] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/13/2023] [Accepted: 07/06/2023] [Indexed: 09/07/2023]
Abstract
Wearable biosensors and smartphone applications can measure physiological variables over multiple days in free-living conditions. We measure food and drink ingestion, glucose dynamics, physical activity, heart rate (HR), and heart rate variability (HRV) in 25 healthy participants over 14 days. We develop a Bayesian inference framework to learn personal parameters that quantify circadian rhythms and physiological responses to external stressors. Modeling the effects of ingestion events on glucose levels reveals that slower glucose decay kinetics elicit larger postprandial glucose spikes, and we uncover a circadian baseline rhythm for glucose with high amplitudes in some individuals. Physical activity and circadian rhythms explain as much as 40%-65% of the HR variance, whereas the variance explained for HRV is more heterogeneous across individuals. A more complex model incorporating activity, HR, and HRV explains up to 15% of additional glucose variability, highlighting the relevance of integrating multiple biosensors to better predict glucose dynamics.
Collapse
Affiliation(s)
- Nicholas E. Phillips
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Nutrition Unit, Service of Endocrinology, Diabetology, Nutrition and Therapeutic Education, Department of Medicine, Geneva University Hospitals (HUG), 1211 Geneva, Switzerland
| | - Tinh-Hai Collet
- Nutrition Unit, Service of Endocrinology, Diabetology, Nutrition and Therapeutic Education, Department of Medicine, Geneva University Hospitals (HUG), 1211 Geneva, Switzerland
- Diabetes Centre, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Felix Naef
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
15
|
Schembri Wismayer D, Laurenti MC, Song Y, Egan AM, Welch AA, Bailey KR, Cobelli C, Dalla Man C, Jensen MD, Vella A. Effects of acute changes in fasting glucose and free fatty acid concentrations on indices of β-cell function and glucose metabolism in subjects without diabetes. Am J Physiol Endocrinol Metab 2023; 325:E119-E131. [PMID: 37285600 PMCID: PMC10393375 DOI: 10.1152/ajpendo.00043.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 06/09/2023]
Abstract
Elevated fasting free fatty acids (FFAs) and fasting glucose are additively associated with impaired glucose tolerance (IGT) and decreased β-cell function [quantified as disposition index (DI)]. We sought to examine how changes in fasting FFA and glucose alter islet function. We studied 10 subjects with normal fasting glucose (NFG) and normal glucose tolerance (NGT) on two occasions. On one occasion, Intralipid and glucose were infused overnight to mimic conditions present in IFG/IGT. In addition, we studied seven subjects with IFG/IGT on two occasions. On one occasion, insulin was infused to lower overnight FFA and glucose concentrations to those observed in people with NFG/NGT. The following morning, a labeled mixed meal was used to measure postprandial glucose metabolism and β-cell function. Elevation of overnight fasting FFA and glucose in NFG/NGT did not alter peak or integrated glucose concentrations (2.0 ± 0.1 vs. 2.0 ± 0.1 Mol per 5 h, Saline vs. Intralipid/glucose, P = 0.55). Although overall β-cell function quantified by the Disposition Index was unchanged, the dynamic component of β-cell responsivity (ϕd) was decreased by Intralipid and glucose infusion (9 ± 1 vs. 16 ± 3 10-9, P = 0.02). In people with IFG/IGT, insulin did not alter postprandial glucose concentrations or indices of β-cell function. Endogenous glucose production and glucose disappearance were also unchanged in both groups. We conclude that acute, overnight changes in FFA, and glucose concentrations do not alter islet function or glucose metabolism in prediabetes.NEW & NOTEWORTHY This experiment studied the effect of changes in overnight concentrations of free fatty acids (FFAs) and glucose on β-cell function and glucose metabolism. In response to elevation of these metabolites, the dynamic component of the β-cell response to glucose was impaired. This suggests that in health overnight hyperglycemia and FFA elevation can deplete preformed insulin granules in the β-cell.
Collapse
Affiliation(s)
- Daniel Schembri Wismayer
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, United States
| | - Marcello C Laurenti
- Biomedical Engineering and Physiology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, United States
| | - Yilin Song
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, United States
| | - Aoife M Egan
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, United States
| | - Andrew A Welch
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, United States
| | - Kent R Bailey
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States
| | - Claudio Cobelli
- Department of Woman and Child's Health, University of Padova, Padova, Italy
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Michael D Jensen
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, United States
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, United States
| |
Collapse
|
16
|
Kovatchev BP, Lobo B. Clinically-Similar Clusters of Daily CGM Profiles: Tracking the Progression of Glycemic Control Over Time. Diabetes Technol Ther 2023. [PMID: 37130300 DOI: 10.1089/dia.2023.0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
BACKGROUND The adoption of CGM results in vast amounts of data, but their interpretation is still more art than exact science. The International Consensus on Time in Range (TIR) proposed the widely accepted TIR system of metrics, which we now take forward by introducing a finite and fixed set of clinically-similar clusters (CSCs), such that the TIR metrics of the daily CGM profiles within a cluster are homogeneous. METHODS CSC definition and validation used 204,710 daily CGM profiles in health, type 1 and type 2 diabetes (T1D, T2D), on different treatments. The CSCs were defined using 23,916 daily CGM profiles (Training set), and the final fixed set of CSCs was obtained using another 37,758 profiles (Validation set). The Testing set (143,036 profiles) was used to establish the robustness and generalizability of the CSCs. RESULTS The final set of CSCs contains 32 clusters. Any daily CGM profile was classifiable to a single CSC which approximated common glycemic metrics of the daily CGM profile, as evidenced by regression analyses with 0 intercept (R-squares≥0.83, e.g., correlation≥0.91), for all TIR and several other metrics. The CSCs distinguished CGM profiles in health, T2D, and T1D on different treatments, and allowed tracking of the daily changes in a person's glycemic control over time. CONCLUSION Daily CGM profiles can be classified into one of 32 prefixed CSCs, which enables a host of applications, e.g. tabulated data interpretation and algorithmic approaches to treatment, database indexing, pattern recognition, and tracking disease progression.
Collapse
Affiliation(s)
- Boris P Kovatchev
- University of Virginia, 2358, Center for Diabetes Technology, Charlottesville, Virginia, United States;
| | - Benjamin Lobo
- University of Virginia, 2358, School of Data Science, Charlottesville, Virginia, United States;
| |
Collapse
|
17
|
Toledo-Marín JQ, Ali T, van Rooij T, Görges M, Wasserman WW. Prediction of Blood Risk Score in Diabetes Using Deep Neural Networks. J Clin Med 2023; 12:jcm12041695. [PMID: 36836230 PMCID: PMC9961355 DOI: 10.3390/jcm12041695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Improving the prediction of blood glucose concentration may improve the quality of life of people living with type 1 diabetes by enabling them to better manage their care. Given the anticipated benefits of such a prediction, numerous methods have been proposed. Rather than attempting to predict glucose concentration, a deep learning framework for prediction is proposed in which prediction is performed using a scale for hypo- and hyper-glycemia risk. Using the blood glucose risk score formula proposed by Kovatchev et al., models with different architectures were trained, including, a recurrent neural network (RNN), a gated recurrent unit (GRU), a long short-term memory (LSTM) network, and an encoder-like convolutional neural network (CNN). The models were trained using the OpenAPS Data Commons data set, comprising 139 individuals, each with tens of thousands of continuous glucose monitor (CGM) data points. The training set was composed of 7% of the data set, while the remaining was used for testing. Performance comparisons between the different architectures are presented and discussed. To evaluate these predictions, performance results are compared with the last measurement (LM) prediction, through a sample-and-hold approach continuing the last known measurement forward. The results obtained are competitive when compared to other deep learning methods. A root mean squared error (RMSE) of 16 mg/dL, 24 mg/dL, and 37 mg/dL were obtained for CNN prediction horizons of 15, 30, and 60 min, respectively. However, no significant improvements were found for the deep learning models compared to LM prediction. Performance was found to be highly dependent on architecture and the prediction horizon. Lastly, a metric to assess model performance by weighing each prediction point error with the corresponding blood glucose risk score is proposed. Two main conclusions are drawn. Firstly, going forward, there is a need to benchmark model performance using LM prediction to enable the comparison between results obtained from different data sets. Secondly, model-agnostic data-driven deep learning models may only be meaningful when combined with mechanistic physiological models; here, it is argued that neural ordinary differential equations may combine the best of both approaches. These findings are based on the OpenAPS Data Commons data set and are to be validated in other independent data sets.
Collapse
Affiliation(s)
- J. Quetzalcóatl Toledo-Marín
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Correspondence:
| | - Taqdir Ali
- Department of Medical Genetics, University of British Columbia, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Tibor van Rooij
- Department of Computer Science, University of British Columbia, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Matthias Görges
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Wyeth W. Wasserman
- Department of Medical Genetics, University of British Columbia, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| |
Collapse
|
18
|
In Silico Evaluation of the Medtronic 780G System While Using the GS3 and Its Calibration-Free Successor, the G4S Sensor. Ann Biomed Eng 2023; 51:211-224. [PMID: 36125605 DOI: 10.1007/s10439-022-03079-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/06/2022] [Indexed: 01/13/2023]
Abstract
In silico simulation studies using 5807 virtual patients with insulin dependent diabetes have been conducted to estimate the risk and efficacy with the closed-loop 780G pump when switching between Medtronic Guardian Sensor 3 (GS3) and Medtronic Guardian 4 Sensor (G4S), next generation calibration free glucose sensor. To demonstrate by utilizing a case study that captures the merits of in silico studies with single hormone insulin dependent virtual patients that include variability in pharmacokinetics/pharmacodynamics, age, gender, insulin sensitivity and BMIs. Also, to show that in silico studies can uniquely isolate the effect of a single variable on clinical outcomes. Simulation studies results were compared to clinical and commercial data and were separated by age groups and pump settings. The commercial data, the clinical study data and the simulation studies predicted that switching between GS3 to G4S will introduce a change in glucose average, percentage time between 70 and 180 mg/dL, and percentage time below 70 mg/dL of: 5.2, 3.4, and 3.1 mg/dL, - 1.1, 0.2, and - 1.1%, and - 0.6, - 1.0, and - 0.3%, respectively. We demonstrated that our simulation studies were able to predict the difference in glycemic outcomes when switching between different sensors in real world setting, better than a small clinical controlled study. As predicted, switching between GS3 and G4S sensors with the 780G system does not introduce clinical risk and maintain the clinical outcomes of the sensor. We demonstrated the ability of insulin dependent diabetes virtual patients to predict clinical outcomes and to augment or even replace some small clinical studies.
Collapse
|
19
|
Sujatha DV. Investigation on Modelling and Identification of Glucose Management system for normal individual and diabetic patient. Comput Chem Eng 2022. [DOI: 10.1016/j.compchemeng.2022.108077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
20
|
Dietrich JW, Dasgupta R, Anoop S, Jebasingh F, Kurian ME, Inbakumari M, Boehm BO, Thomas N. SPINA Carb: a simple mathematical model supporting fast in-vivo estimation of insulin sensitivity and beta cell function. Sci Rep 2022; 12:17659. [PMID: 36271244 PMCID: PMC9587026 DOI: 10.1038/s41598-022-22531-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 10/17/2022] [Indexed: 01/18/2023] Open
Abstract
Modelling insulin-glucose homeostasis may provide novel functional insights. In particular, simple models are clinically useful if they yield diagnostic methods. Examples include the homeostasis model assessment (HOMA) and the quantitative insulin sensitivity check index (QUICKI). However, limitations of these approaches have been criticised. Moreover, recent advances in physiological and biochemical research prompt further refinement in this area. We have developed a nonlinear model based on fundamental physiological motifs, including saturation kinetics, non-competitive inhibition, and pharmacokinetics. This model explains the evolution of insulin and glucose concentrations from perturbation to steady-state. Additionally, it lays the foundation of a structure parameter inference approach (SPINA), providing novel biomarkers of carbohydrate homeostasis, namely the secretory capacity of beta-cells (SPINA-GBeta) and insulin receptor gain (SPINA-GR). These markers correlate with central parameters of glucose metabolism, including average glucose infusion rate in hyperinsulinemic glucose clamp studies, response to oral glucose tolerance testing and HbA1c. Moreover, they mirror multiple measures of body composition. Compared to normal controls, SPINA-GR is significantly reduced in subjects with diabetes and prediabetes. The new model explains important physiological phenomena of insulin-glucose homeostasis. Clinical validation suggests that it may provide an efficient biomarker panel for screening purposes and clinical research.
Collapse
Affiliation(s)
- Johannes W. Dietrich
- grid.5570.70000 0004 0490 981XDiabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef Hospital, Ruhr University Bochum, NRW, Gudrunstr. 56, 44791 Bochum, Germany ,Diabetes Centre Bochum-Hattingen, St. Elisabeth-Hospital Blankenstein, Im Vogelsang 5-11, 45527 Hattingen, NRW Germany ,grid.5570.70000 0004 0490 981XCentre for Rare Endocrine Diseases, Ruhr Centre for Rare Diseases (CeSER), Ruhr University Bochum and Witten/Herdecke University, Alexandrinenstr. 5, 44791 Bochum, NRW Germany ,Centre for Diabetes Technology, Catholic Hospitals Bochum, Gudrunstr. 56, 44791 Bochum, NRW, Germany
| | - Riddhi Dasgupta
- grid.11586.3b0000 0004 1767 8969Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, 632004 India
| | - Shajith Anoop
- grid.11586.3b0000 0004 1767 8969Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, 632004 India
| | - Felix Jebasingh
- grid.11586.3b0000 0004 1767 8969Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, 632004 India
| | - Mathews E. Kurian
- grid.11586.3b0000 0004 1767 8969Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, 632004 India
| | - Mercy Inbakumari
- grid.11586.3b0000 0004 1767 8969Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, 632004 India
| | - Bernhard O. Boehm
- grid.59025.3b0000 0001 2224 0361Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore, 308232 Singapore ,grid.6582.90000 0004 1936 9748Department of Internal Medicine I, Ulm University Medical Centre, Ulm University, 89070 Ulm, Germany ,grid.240988.f0000 0001 0298 8161Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Nihal Thomas
- grid.11586.3b0000 0004 1767 8969Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, 632004 India
| |
Collapse
|
21
|
Universal Minimal Model for Glucose-Insulin Relationship with the Influence of Food Dynamic. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:8990767. [PMID: 36203526 PMCID: PMC9532137 DOI: 10.1155/2022/8990767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/29/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022]
Abstract
We propose a minimal model defining the relationship between glucose and insulin with the added influence of food intakes. The constructed model consists of a system of 3 nonlinear ordinary differential equations (ODEs). The solutions of our model for both normal and diabetic subjects are compared with a minimal model and a maximal model representing the same relationship. We found that the outputs of our model are similar to those from the minimal and maximal models for both normal and diabetic subjects; the R2 are 0.9997 and 0.9922, respectively, when compared with the minimal model, and are 0.9995 and 0.9940, respectively, when compared with the maximal model. Moreover, the relative errors between solutions are at most 0.9035% and as low as 1.488 × 10−2% on average when compared with the minimal model for normal subjects and at most 1.331% and as low as 0.1159% on average for diabetic subjects. The discrepancy between our model and the maximal model are at most 1.590% and 5.453% for normal and diabetic subjects, respectively, with a relative error averaging 0.2138% and 0.9002% for normal and diabetic subjects, respectively.
Collapse
|
22
|
Subramanian V, Bagger JI, Holst JJ, Knop FK, Vilsbøll T. A glucose-insulin-glucagon coupled model of the isoglycemic intravenous glucose infusion experiment. Front Physiol 2022; 13:911616. [PMID: 36148302 PMCID: PMC9485803 DOI: 10.3389/fphys.2022.911616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Type 2 diabetes (T2D) is a pathophysiology that is characterized by insulin resistance, beta- and alpha-cell dysfunction. Mathematical models of various glucose challenge experiments have been developed to quantify the contribution of insulin and beta-cell dysfunction to the pathophysiology of T2D. There is a need for effective extended models that also capture the impact of alpha-cell dysregulation on T2D. In this paper a delay differential equation-based model is developed to describe the coupled glucose-insulin-glucagon dynamics in the isoglycemic intravenous glucose infusion (IIGI) experiment. As the glucose profile in IIGI is tailored to match that of a corresponding oral glucose tolerance test (OGTT), it provides a perfect method for studying hormone responses that are in the normal physiological domain and without the confounding effect of incretins and other gut mediated factors. The model was fit to IIGI data from individuals with and without T2D. Parameters related to glucagon action, suppression, and secretion as well as measures of insulin sensitivity, and glucose stimulated response were determined simultaneously. Significant impairment in glucose dependent glucagon suppression was observed in patients with T2D (duration of T2D: 8 (6-36) months) relative to weight matched control subjects (CS) without diabetes (k1 (mM)-1: 0.16 ± 0.015 (T2D, n = 7); 0.26 ± 0.047 (CS, n = 7)). Insulin action was significantly lower in patients with T2D (a1 (10 pM min)-1: 0.000084 ± 0.0000075 (T2D); 0.00052 ± 0.00015 (CS)) and the Hill coefficient in the equation for glucose dependent insulin response was found to be significantly different in T2D patients relative to CS (h: 1.4 ± 0.15; 1.9 ± 0.14). Trends in parameters with respect to fasting plasma glucose, HbA1c and 2-h glucose values are also presented. Significantly, a negative linear relationship is observed between the glucagon suppression parameter, k1, and the three markers for diabetes and is thus indicative of the role of glucagon in exacerbating the pathophysiology of diabetes (Spearman Rank Correlation: (n = 12; (-0.79, 0.002), (-0.73,.007), (-0.86,.0003)) respectively).
Collapse
Affiliation(s)
- Vijaya Subramanian
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Jonatan I. Bagger
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Jens J. Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Filip K. Knop
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Cobelli C, Dalla Man C. Minimal and Maximal Models to Quantitate Glucose Metabolism: Tools to Measure, to Simulate and to Run in Silico Clinical Trials. J Diabetes Sci Technol 2022; 16:1270-1298. [PMID: 34032128 PMCID: PMC9445339 DOI: 10.1177/19322968211015268] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Several models have been proposed to describe the glucose system at whole-body, organ/tissue and cellular level, designed to measure non-accessible parameters (minimal models), to simulate system behavior and run in silico clinical trials (maximal models). Here, we will review the authors' work, by putting it into a concise historical background. We will discuss first the parametric portrait provided by the oral minimal models-building on the classical intravenous glucose tolerance test minimal models-to measure otherwise non-accessible key parameters like insulin sensitivity and beta-cell responsivity from a physiological oral test, the mixed meal or the oral glucose tolerance tests, and what can be gained by adding a tracer to the oral glucose dose. These models were used in various pathophysiological studies, which we will briefly review. A deeper understanding of insulin sensitivity can be gained by measuring insulin action in the skeletal muscle. This requires the use of isotopic tracers: both the classical multiple-tracer dilution and the positron emission tomography techniques are discussed, which quantitate the effect of insulin on the individual steps of glucose metabolism, that is, bidirectional transport plasma-interstitium, and phosphorylation. Finally, we will present a cellular model of insulin secretion that, using a multiscale modeling approach, highlights the relations between minimal model indices and subcellular secretory events. In terms of maximal models, we will move from a parametric to a flux portrait of the system by discussing the triple tracer meal protocol implemented with the tracer-to-tracee clamp technique. This allows to arrive at quasi-model independent measurement of glucose rate of appearance (Ra), endogenous glucose production (EGP), and glucose rate of disappearance (Rd). Both the fast absorbing simple carbs and the slow absorbing complex carbs are discussed. This rich data base has allowed us to build the UVA/Padova Type 1 diabetes and the Padova Type 2 diabetes large scale simulators. In particular, the UVA/Padova Type 1 simulator proved to be a very useful tool to safely and effectively test in silico closed-loop control algorithms for an artificial pancreas (AP). This was the first and unique simulator of the glucose system accepted by the U.S. Food and Drug Administration as a substitute to animal trials for in silico testing AP algorithms. Recent uses of the simulator have looked at glucose sensors for non-adjunctive use and new insulin molecules.
Collapse
Affiliation(s)
- Claudio Cobelli
- Department of Woman and Child’s Health University of Padova, Padova, Italy
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padova, Italy
| |
Collapse
|
24
|
Visentin R, Cobelli C, Dalla Man C. A software interface for in silico testing of type 2 diabetes treatments. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 223:106973. [PMID: 35792365 DOI: 10.1016/j.cmpb.2022.106973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 06/09/2022] [Accepted: 06/21/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND OBJECTIVE The increasing incidence of diabetes continuously stimulates the research on new antidiabetic drugs. Computer simulation can save time and costs, alleviating the need of animal trials and providing useful information for optimal experiment design and drug dosing. We recently presented a type 2 diabetes (T2D) simulator as tool for in silico testing of new molecules and guiding treatment optimization. Here we present a user-friendly interface aimed to increase the usability of the simulator. METHOD The simulator, based on a large-scale glucose, insulin, and C-peptide model and equipped with 100 virtual subjects well describing system dynamics in a real T2D population, is extended to incorporate pharmacokinetics/pharmacodynamics (PK/PD) of a drug of interest. A graphical interface is developed on top of the simulator, allowing an easy design of in silico experiments: specifically, it is possible to select the population size to test, design the experiment (crossover or parallel), its duration and the sampling grid, choose glucose and insulin doses, and define treatment PK/PD and dose administered. The simulator also provides the outcome metrics requested by the user, and performs statistical comparisons among treatments and/or placebo. RESULTS To illustrate the potential of the simulator, we provided a case study using metformin and liraglutide. Literature-based PK/PD models of metformin and liraglutide have been incorporated in the simulator, by modulating key drug-sensitive model parameters. An in silico placebo-controlled trial has been done by simulating a three-arm meal tolerance test with subjects receiving placebo, metformin 850 mg, liraglutide 1.80 mg, respectively. The obtained results are in agreement with the clinical evidences, in terms of main glucose, insulin, and C-peptide outcome metrics. CONCLUSIONS We developed a user-friendly software interface for the T2D simulator to support the design and test of new antidiabetic drugs and treatments. This increases the simulator usability, making it suitable also for users who have low experience with computer programming.
Collapse
Affiliation(s)
- Roberto Visentin
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Claudio Cobelli
- Department of Woman and Child's Health, University of Padova, Padova, Italy
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padova, Italy.
| |
Collapse
|
25
|
Computational-Model-Based Biopharmaceutics for p53 Pathway Using Modern Control Techniques for Cancer Treatment. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12115748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The p53 pathway has been the focus of many researchers in the last few decades owing to its pivotal role as a frontline cancer suppressant protein. It plays a vital role in maintaining cell cycle checkpoints and cell apoptosis in response to a broken DNA strand. This is why it is found in the mutated form in more than 50% of malignant tumors. To overcome this, various drugs have been proposed to revive the p53 pathway in cancer patients. Small-molecule-based drugs, such as Nutlin 3a, which are capable of performing this stimulation, are at the fore of advanced clinical trials. However, the calculation of their dosage is a challenge. In this work, a method to determine the dosage of Nutlin 3a is investigated. A control-systems-based model is developed to study the response of the wild-type p53 protein to this drug. The proposed strategy regulates the p53 protein along with negative and positive feedback loops mediated by the MDM2 and MDM2 mRNA, respectively, along with the reversible repression of MDM2 caused by Nutlin 3a. For a broader perspective, the reported PBK dynamics of Nutlin 3a are also incorporated. It has been reported that p53 responds to stresses in two ways in terms of concentration to this drug: either it is a sustained (constant) or an oscillatory response. The claimed dosage strategy turned out to be appropriate for sustained p53 response. However, for the induction of oscillations, inhibition of MDM2 is not enough; rather, anti-repression of the p53–MDM2 complex is also needed, which opens new horizons for a new drug design paradigm.
Collapse
|
26
|
Lim MH, Cho YM, Kim S. Multi-task disentangled autoencoder for time-series data in glucose dynamics. IEEE J Biomed Health Inform 2022; 26:4702-4713. [PMID: 35588418 DOI: 10.1109/jbhi.2022.3175928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The objective of this study is to propose MD-VAE: a multi-task disentangled variational autoencoders (VAE) for exploring characteristics of latent representations (LR) and exploiting LR for diverse tasks including glucose forecasting, event detection, and temporal clustering.
Collapse
|
27
|
Sharma A, Singh HP, Nilam. A methodical survey of mathematical model-based control techniques based on open and closed loop control approach for diabetes management. INT J BIOMATH 2022. [DOI: 10.1142/s1793524522500516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Disturbance of blood sugar level is controlled through well-known biomechanical feedback loops: high levels of glucose in blood facilitate to release insulin from the pancreas which accelerates the absorption rate of cellular glucose. Low glucose levels encourage to release pancreatic glucagon which induces glycogen breakdown to glucose in the liver. These bio-control systems do not function properly in diabetic patients. Though the control of disease seems intuitively easy, in real life, due to many differences in structure by diet and fasting, exercise, medications, patient’s profile and other stressors, it is not that easy. The mathematical models of the glucose-insulin regulatory system follow the patient’s physiological conditions which make it difficult to identify and estimate all the model parameters. In this paper, we have given a systematic literature review on mathematical models of the diabetic patients, and various kinds of disease control techniques through the development of open and closed loop insulin deliver command system and optimization of exogenous insulin rate. It demonstrates the open and closed loop type model-based control strategies underlying the assumptions of the concerned models. The combination of mathematical model with control strategies such as genetic algorithm (GA), neural network (NN), sliding mode controller (SMC), model predictive controller (MPC), and fuzzy logic control (FLC) has been considered, which provides an overview of this area, highlighting the control profile over the diabetic model with promising clinical results, outlining key challenges, and identifying needs for the future research. Also, the significance of these control algorithms has been discussed in the presence of the noises, the controller’s robustness and various other disturbances. It provides substantial information on diabetes management through various control techniques.
Collapse
Affiliation(s)
- Ankit Sharma
- Department of Applied Mathematics, Delhi Technological University, Delhi 110042, India
| | | | - Nilam
- Department of Applied Mathematics, Delhi Technological University, Delhi 110042, India
| |
Collapse
|
28
|
Builes-Montaño CE, Lema-Perez L, Garcia-Tirado J, Alvarez H. Main glucose hepatic fluxes in healthy subjects predicted from a phenomenological-based model. Comput Biol Med 2022; 142:105232. [DOI: 10.1016/j.compbiomed.2022.105232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/08/2022] [Accepted: 01/09/2022] [Indexed: 11/28/2022]
|
29
|
Gautier T, Silwal R, Saremi A, Boss A, Breton MD. Modeling the Effect of Subcutaneous Lixisenatide on Glucoregulatory Endocrine Secretions and Gastric Emptying in Type 2 Diabetes to Simulate the Effect of iGlarLixi Administration Timing on Blood Sugar Profiles. J Diabetes Sci Technol 2022; 16:428-433. [PMID: 34013770 PMCID: PMC8847729 DOI: 10.1177/19322968211015671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND As type 2 diabetes (T2D) progresses, intensification to combination therapies, such as iGlarLixi (a fixed-ratio GLP-1 RA and basal insulin combination), may be required. Here a simulation study was used to assess the effect of iGlarLixi administration timing (am vs pm) on blood sugar profiles. METHODS Models of lixisenatide were built with a selection procedure, optimizing measurement fits and model complexity, and were included in a pre-existing T2D simulation platform containing glargine models. With the resulting tool, a simulated trial was conducted with 100 in-silico participants with T2D. Individuals were given iGLarLixi either before breakfast or before an evening meal for 2 weeks and daily glycemic profiles were analyzed. In the model, breakfast was considered the largest meal of the day. RESULTS A similar percentage of time within 24 hours was spent with blood sugar levels between 70 to 180 mg/dL when iGlarLixi was administered pre-breakfast or pre-evening meal (73% vs 71%, respectively). Overall percent of time with blood glucose levels above 180 mg/dL within a 24-hour period was similar when iGlarLixi was administered pre-breakfast or pre-evening meal (26% vs 28%, respectively). Rates of hypoglycemia were low in both regimens, with a blood glucose concentration of below 70 mg/dL only observed for 1% of the 24-hour time period for either timing of administration. CONCLUSIONS Good efficacy was observed when iGlarlixi was administered pre-breakfast; however, administration of iGlarlixi pre-evening meal was also deemed to be effective, even though in the model the size of the evening meal was smaller than that of the breakfast.
Collapse
Affiliation(s)
- Thibault Gautier
- Center for Diabetes Technology, University of
Virginia, Charlottesville, VA, USA
| | - Rupesh Silwal
- Center for Diabetes Technology, University of
Virginia, Charlottesville, VA, USA
| | | | - Anders Boss
- Medical Affairs, Sanofi, Bridgewater, NJ,
USA
| | - Marc D. Breton
- Center for Diabetes Technology, University of
Virginia, Charlottesville, VA, USA
| |
Collapse
|
30
|
olde Scheper TV. Controlled bio-inspired self-organised criticality. PLoS One 2022; 17:e0260016. [PMID: 35073308 PMCID: PMC8786161 DOI: 10.1371/journal.pone.0260016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/29/2021] [Indexed: 11/26/2022] Open
Abstract
Complex biological systems are considered to be controlled using feedback mechanisms. Reduced systems modelling has been effective to describe these mechanisms, but this approach does not sufficiently encompass the required complexity that is needed to understand how localised control in a biological system can provide global stable states. Self-Organised Criticality (SOC) is a characteristic property of locally interacting physical systems, which readily emerges from changes to its dynamic state due to small nonlinear perturbations. These small changes in the local states, or in local interactions, can greatly affect the total system state of critical systems. It has long been conjectured that SOC is cardinal to biological systems, that show similar critical dynamics, and also may exhibit near power-law relations. Rate Control of Chaos (RCC) provides a suitable robust mechanism to generate SOC systems, which operates at the edge of chaos. The bio-inspired RCC method requires only local instantaneous knowledge of some of the variables of the system, and is capable of adapting to local perturbations. Importantly, connected RCC controlled oscillators can maintain global multi-stable states, and domains where power-law relations may emerge. The network of oscillators deterministically stabilises into different orbits for different perturbations, and the relation between the perturbation and amplitude can show exponential and power-law correlations. This can be considered to be representative of a basic mechanism of protein production and control, that underlies complex processes such as homeostasis. Providing feedback from the global state, the total system dynamic behaviour can be boosted or reduced. Controlled SOC can provide much greater understanding of biological control mechanisms, that are based on distributed local producers, with remote consumers of biological resources, and globally defined control.
Collapse
Affiliation(s)
- Tjeerd V. olde Scheper
- School of Engineering, Computing and Mathematics, Oxford Brookes University, Wheatley Campus, Oxford, United Kingdom
| |
Collapse
|
31
|
Ahdab MA, Leth J, Knudsen T, Vestergaard P, Clausen HG. Glucose-insulin mathematical model for the combined effect of medications and life style of Type 2 diabetic patients. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2021.108170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
32
|
Lopez-Zazueta C, Stavdahl O, Fougner AL. Low-Order Nonlinear Animal Model of Glucose Dynamics for a Bihormonal Intraperitoneal Artificial Pancreas. IEEE Trans Biomed Eng 2021; 69:1273-1280. [PMID: 34748476 DOI: 10.1109/tbme.2021.3125839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The design of an Artificial Pancreas to regulate blood glucose levels requires reliable control methods. Model Predictive Control has emerged as a promising approach for glycemia control. However, model-based control methods require computationally simple and identifiable mathematical models that represent glucose dynamics accurately, which is challenging due to the complexity of glucose homeostasis. METHODS In this work, a simple model is deduced to estimate blood glucose concentration in subjects with Type 1 Diabetes Mellitus. Novel features in the model are power-law kinetics for intraperitoneal insulin absorption and a separate glucagon sensitivity state. Profile likelihood and a method based on singular value decomposition of the sensitivity matrix are carried out to assess parameter identifiability and guide a model reduction for improving the identification of parameters. RESULTS A reduced model with 10 parameters is obtained and calibrated, showing good fit to experimental data from pigs where insulin and glucagon boluses were delivered in the intraperitoneal cavity. CONCLUSION A simple model with power-law kinetics can accurately represent glucose dynamics submitted to intraperitoneal insulin and glucagon injections. IMPORTANCE The parameters of the reduced model were not found to lack of local practical or structural identifiability.
Collapse
|
33
|
Visentin R, De Lazzari M. A Novel Method for Generation of In Silico Subjects with Type 2 Diabetes. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2021; 2021:1380-1383. [PMID: 34891542 DOI: 10.1109/embc46164.2021.9629678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
A type 2 diabetes (T2D) simulator has been recently proposed for supporting drug development and treatment optimization. This tool consists of a physiological model of glucose/insulin/C-peptide dynamics and a virtual cohort of T2D subjects (i.e., random extractions of model parameterizations from a joint parameter distribution) well describing both average and variability realistic T2D dynamics . However, the state-of-art procedure to get a reliable virtual population requires some post-processing after subject extraction, in order to discard implausible behaviors. We propose an improved method for virtual subjects' generation to overcome this burdensome task. To do so, we first assessed a refined joint parameter distribution, from which extracting a number of subjects, greater than the target population size. Then, target-size subsets are undersampled from the large cohort. The final virtual population is selected among the subsets as the one maximizing the similarity with T2D data and model parameter distribution, by means of measurement' outcome metrics and Euclidian distance (Δ), respectively. In the final population, almost all the outcome metrics are statistically identical to the clinical counterparts (p-value>0.05) and model parameters' distribution differs by ~5-10% from that derived from data. The methodology described here is flexible, thus resulting suitable for different T2D stages and type 1 diabetes, as well.Clinical Relevance- A straightforward subjects' generation would ease the availability of tailored in silico trials for testing diabetes treatment in a specific population.
Collapse
|
34
|
Hughes J, Gautier T, Colmegna P, Fabris C, Breton MD. Replay Simulations with Personalized Metabolic Model for Treatment Design and Evaluation in Type 1 Diabetes. J Diabetes Sci Technol 2021; 15:1326-1336. [PMID: 33218280 PMCID: PMC8655285 DOI: 10.1177/1932296820973193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND The capacity to replay data collected in real life by people with type 1 diabetes mellitus (T1DM) would lead to individualized (vs population) assessment of treatment strategies to control blood glucose and possibly true personalization. Patek et al introduced such a technique, relying on regularized deconvolution of a population glucose homeostasis model to estimate a residual additive signal and reproduce the experimental data; therefore, allowing the subject-specific replay of what-if scenarios by altering the model inputs (eg, insulin). This early method was shown to have a limited domain of validity. We propose and test in silico a similar approach and extend the method applicability. METHODS A subject-specific model personalization of insulin sensitivity and meal-absorption parameters is performed. The University of Virginia (UVa)/Padova T1DM simulator is used to generate experimental scenarios and test the ability of the methodology to accurately reproduce changes in glucose concentration to alteration in meal and insulin inputs. Method performance is assessed by comparing true (UVa/Padova simulator) and replayed glucose traces, using the mean absolute relative difference (MARD) and the Clarke error grid analysis (CEGA). RESULTS Model personalization led to a 9.08 and 6.07 decrease in MARD over a prior published method of replaying altered insulin scenarios for basal and bolus changes, respectively. Replay simulations achieved high accuracy, with MARD <10% and more than 95% of readings falling in the CEGA A-B zones for a wide range of interventions. CONCLUSIONS In silico studies demonstrate that the proposed method for replay simulation is numerically and clinically valid over broad changes in scenario inputs, indicating possible use in treatment optimization.
Collapse
Affiliation(s)
- Jonathan Hughes
- Center for Diabetes Technology,
University of Virginia, Charlottesville, VA, USA
| | - Thibault Gautier
- Center for Diabetes Technology,
University of Virginia, Charlottesville, VA, USA
| | - Patricio Colmegna
- Center for Diabetes Technology,
University of Virginia, Charlottesville, VA, USA
| | - Chiara Fabris
- Center for Diabetes Technology,
University of Virginia, Charlottesville, VA, USA
| | - Marc D Breton
- Center for Diabetes Technology,
University of Virginia, Charlottesville, VA, USA
- Marc D Breton, PhD, Center for Diabetes
Technology, University of Virginia, 560 Ray C Hunt Dr, Charlottesville, VA
22903, USA.
| |
Collapse
|
35
|
Batmani Y, Khodakaramzadeh S, Moradi P. Automatic Artificial Pancreas Systems Using an Intelligent Multiple-Model PID Strategy. IEEE J Biomed Health Inform 2021; 26:1708-1717. [PMID: 34587104 DOI: 10.1109/jbhi.2021.3116376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In this paper, an individualized intelligent multiple-model technique is proposed to design automatic artificial pancreas (AP) systems for the glycemic regulation of type 1 diabetic patients. At first, using the multiple-model concept, the insulin-glucose regulatory system is mathematically identified by constructing some local models. In this step, trade-offs between the number of local models and the complexity of the overall closed-loop system are made by defining and solving a bi-objective optimization problem. Then, optimal AP systems are designed by tuning a bank of proportionalintegralderivative (PID) controllers via the genetic algorithm (GA). A fuzzy gain scheduling strategy is employed to determine the participation percentages of the PID controllers in the control action. Finally, two safety mechanisms, called insulin on board (IOB) constraint and pump shut-off, are installed in the AP systems to enhance their performance. To assess the proposed AP systems, in silico experiments are performed on virtual patients of the UVA/Padova metabolic simulator. The obtained results reveal that the proposed intelligent multiple-model methodology leads to AP systems with limited hyperglycemia and no severe hypoglycemia.
Collapse
|
36
|
Wang W, Wang S, Geng Y, Qiao Y, Wu T. An OGI model for personalized estimation of glucose and insulin concentration in plasma. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:8499-8523. [PMID: 34814309 DOI: 10.3934/mbe.2021420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Plasma glucose concentration (PGC) and plasma insulin concentration (PIC) are two essential metrics for diabetic regulation, but difficult to be measured directly. Often, PGC and PIC are estimated from continuous glucose monitoring and insulin delivery data. Nevertheless, the inter-individual variability and external disturbance (e.g. carbohydrate intake) bring challenges for accurate estimations. This study is to estimate PGC and PIC adaptively by identifying personalized parameters and external disturbances. An observable glucose-insulin (OGI) dynamic model is established to describe insulin absorption, glucose regulation, and glucose transport. The model parameters and disturbances can be extended to observable state variables and be identified dynamically by Bayesian filtering estimators. Two basic Gaussian noise based Bayesian filtering estimators, extended Kalman filtering (EKF) and unscented Kalman filtering (UKF), are implemented. Recognizing the prevalence of non-Gaussian noise, in this study, two new filtering estimators: particle filtering with Gaussian noise (PFG), and particle filtering with mixed non-Gaussian noise (PFM) are designed and implemented. The proposed OGI model in conjunction with the estimators is evaluated using the data from 30 in-silico subjects and 10 human participants. For in-silico subjects, the OGI with PFM estimator has the ability to estimate PIC and PGC adaptively, achieving RMSE of PIC 9.49±3.81 mU/L, and PGC 0.89±0.19 mmol/L. For human, the OGI with PFM has the promise to identify disturbances (95.46%±0.65% accurate rate of meal identification). OGI model provides a way to fully personalize the parameters and external disturbances in real time, and has potential clinical utility for artificial pancreas.
Collapse
Affiliation(s)
- Weijie Wang
- School of Automation Science and Electrical Engineering, Beihang University, Beijing 100191, China
| | - Shaoping Wang
- School of Automation Science and Electrical Engineering, Beihang University, Beijing 100191, China
- Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Beihang University, Beijing 100191, China
| | - Yixuan Geng
- School of Automation Science and Electrical Engineering, Beihang University, Beijing 100191, China
| | - Yajing Qiao
- School of Automation Science and Electrical Engineering, Beihang University, Beijing 100191, China
| | - Teresa Wu
- School of Computing, Informatics, and Decision Systems Engineering, Arizona State University and College of Medicine, Mayo Clinic, Tempe AZ 85281, the USA
| |
Collapse
|
37
|
Faccioli S, Facchinetti A, Sparacino G, Pillonetto G, Del Favero S. Linear Model Identification for Personalized Prediction and Control in Diabetes. IEEE Trans Biomed Eng 2021; 69:558-568. [PMID: 34347589 DOI: 10.1109/tbme.2021.3101589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Type-1 diabetes (T1D) is a metabolic disease, characterized by impaired blood glucose (BG) regulation, which forces patients to multiple daily therapeutic actions, the most critical of which is exogenous insulin administration. T1D management can considerably benefit of mathematical models enabling accurate BG predictions and effective/safe automated insulin delivery. In building these models, dealing with large inter- and intra-patient variability in glucose-insulin dynamics represents a major challenge. The aim of the present work is to assess linear black-box methods, including a novel non-parametric methodology, for learning individualized models of glucose response to insulin and meal, suitable for model-based prediction and control. METHODS We focus on data-driven techniques for linear model-learning and compare the state-of-art parametric pipeline, exploring all its degrees of freedom (including population vs. individualized parameter identification, model class chosen among ARX/ARMAX/ARIMAX/Box-Jenkins, model order selection criteria, etc.), with a novel non-parametric approach based on Gaussian regression and stable spline kernel. By using data collected in 11 T1D individuals, we evaluate effectiveness of the different models by measuring root mean squared error (RMSE), coefficient of determination (COD), and time gain of the associated BG predictors. RESULTS Among the tested approaches, the non-parametric technique results in the best prediction performance: median RMSE=29.8mg/dL, and median COD=57.4%, for a prediction horizon (PH) of 60 min. With respect to the state-of-the-art parametric techniques, the non-parametric approach grants a COD improvement of about 2%, 7%, 21%, and 41% for PH = 30, 60, 90, and 120 min (paired-sample t-test p 0.001, p=0.003, p=0.03, and p=0.07 respectively). CONCLUSION Non-parametric linear model-learning grants statistically significant improvement with respect to the state-of-art parametric approach. The higher PH, the more pronounced the improvement. SIGNIFICANCE The use of a linear non-parametric model-learning approach for model-based prediction and control could bring to a more prompt, safe and effective T1D management.
Collapse
|
38
|
Homayounzade M. Variable structure robust controller design for blood glucose regulation for type 1 diabetic patients: A backstepping approach. IET Syst Biol 2021; 15:173-183. [PMID: 34236138 PMCID: PMC8675804 DOI: 10.1049/syb2.12032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 11/20/2022] Open
Abstract
Diabetes mellitus type 1 occurs when β - cells in the pancreas are destroyed by the immune system. As a result, the pancreas cannot produce adequate insulin, and the glucose enters the cells to produce energy. To elevate the glycaemic concentration, sufficient amount of insulin should be taken orally or injected into the human body. Artificial pancreas is a device that automatically regulates the level of body insulin by injecting the requisite amount of insulin into the human body. A finite-time robust feedback controller based on the Extended Bergman Minimal Model is designed here. The controller is designed utilizing the backstepping approach and is robust against the unknown external disturbance and parametric uncertainties. The stability of the system is proved using the Lyapunov theorem. The controller is exponentially stable and hence provides the finite-time convergence of the blood glucose concentration to its desired magnitude. The effectiveness of the proposed control method is shown through simulation in MATLAB/Simulink environment via comparisons with previous studies.
Collapse
|
39
|
Fakhroleslam M, Bozorgmehry Boozarjomehry R. A multi‐objective optimal insulin bolus advisor for type 1 diabetes based on personalized model and daily diet. ASIA-PAC J CHEM ENG 2021. [DOI: 10.1002/apj.2651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Mohammad Fakhroleslam
- Process Engineering Department, Faculty of Chemical Engineering Tarbiat Modares University Tehran Iran
| | | |
Collapse
|
40
|
Eberle C, Ament C. A combined in vivo and in silico model shows specific predictors of individual trans-generational diabetic programming. J Dev Orig Health Dis 2021; 12:396-403. [PMID: 32808917 DOI: 10.1017/s2040174420000471] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Diabetic pregnancies are cleary associated with maternal type 2 diabetes and metabolic syndrome as well as atherosclerotic diseases in the offspring. The global prevalence of hyperglycemia in pregnancy was estimated as 15.8% of live births to women in 2019, with an upward trend. Numerous parental risk factors as well as trans-generational mechanisms targeting the utero-placental system, leading to diabetes, dysmetabolic and atherosclerotic conditions in the next generation, seem to be involved within this pathophysiological context. To focus on the predictable impact of trans-generational diabetic programming, we studied age- and gender-matched offspring of diabetic and nondiabetic mothers. The offspring generation consists of three groups: C57BL/6-J-Ins2Akita (positive control group), wild-type C57BL/6-J-Ins2Akita (experimental group), and C57BL/6-J mice (negative control group). We undertook intraperitoneal glucose tolerance tests at 3 and 11 weeks of age. Moreover, this in vivo model was complemented by a corresponding in silico model. Although at 3 weeks of age, no significant effects could be observed, we could demonstrate at 11 weeks of age characteristic and significant differences in relation to maternal diabetic imprinting based on the in silico model-based predictors. These predictors allow the generation of a concise classification tree assigning maternal diabetic imprinting correctly in 91% of study cases. Our data show that hyperglycemic in utero milieu contributes to trans-generational diabetic programming leading to impaired glucose tolerance in the offspring of diabetic mothers early on. These observations can be clearly and early distinguished from genetically determined diabetes, for example, type 1 diabetes, in which basal glucose values are significantly raised.
Collapse
Affiliation(s)
- Claudia Eberle
- Hochschule Fulda - University of Applied Sciences, Medicine with Specialization in Internal Medicine and General Medicine, 36037Fulda, Germany
- Diabetes Center and Department of Internal Medicine IV of the Ludwig-Maximilians University of Munich (LMU), 80336München, Germany
| | - Christoph Ament
- Chair of Control Engineering, University Augsburg, 86159Augsburg, Germany
| |
Collapse
|
41
|
Grosman B, Wu D, Parikh N, Roy A, Voskanyan G, Kurtz N, Sturis J, Cohen O, Ekelund M, Vigersky R. Fast-acting insulin aspart (Fiasp®) improves glycemic outcomes when used with MiniMed TM 670G hybrid closed-loop system in simulated trials compared to NovoLog®. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2021; 205:106087. [PMID: 33873075 DOI: 10.1016/j.cmpb.2021.106087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 03/30/2021] [Indexed: 06/12/2023]
Abstract
INTRODUCTION Medtronic has developed a virtual patient simulator for modeling and predicting insulin therapy outcomes for people with type 1 diabetes (T1D). An enhanced simulator was created to estimate outcomes when using the MiniMedTM 670G system with standard NovoLog® (EU: NovoRapid, US: NovoLog) versus Fiasp ® by using clinical data. METHODS Sixty-seven participants' PK profiles were generated per type of insulin (Total of 134 PK profiles). 7,485 virtual patients' PK measurements was matched with one of the 67 NovoLog® PK Tmax values. These 7,485 virtual patients were then simulated using the Medtronic MiniMed™ 670G algorithm following an in-silico protocol of 90 days: 14 days in open loop (Manual Mode) followed by 76 days in closed loop (Auto Mode). Simulation study was repeated with each NovoLog® PK profile being replaced by its corresponding Fiasp® PK profile in the same virtual patient. To validate our in-silico analysis, we compared the results of "actual" 19 "real life" patients from a clinical study RESULTS: Simulated overall and postprandial glycemic outcomes improved in all age groups with Fiasp®. The percentage of time in the euglycemic range increased by about ~2.2% with Fiasp®, in all age groups (p < 0.01). The percentage of time spent at <70 mg/dL was reduced by about ~0.6% with insulin Fiasp® (p < 0.01) and the mean glucose was reduced by about 1.3 mg/dL (p < 0.01), excluding those age <7 years. The simulated mean postprandial SG was reduced by ~5 mg/dL, a significant difference for all age groups. A clinical study results showed similar improvements with MiniMedTM 670G system when switching from NovoLog® to Fiasp®. CONCLUSIONS The simulation studies indicate that using Fiasp® in place of NovoLog® with the MiniMedTM 670G system will significantly improve the time spent in the healthy, euglycemic range and reduce exposure to hyperglycemia and hypoglycemia in most patients.
Collapse
Affiliation(s)
| | - Di Wu
- Medtronic Diabetes, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Probabilistic Model of Transition between Categories of Glucose Profiles in Patients with Type 1 Diabetes Using a Compositional Data Analysis Approach. SENSORS 2021; 21:s21113593. [PMID: 34064157 PMCID: PMC8196791 DOI: 10.3390/s21113593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 01/06/2023]
Abstract
The time spent in glucose ranges is a common metric in type 1 diabetes (T1D). As the time in one day is finite and limited, Compositional Data (CoDa) analysis is appropriate to deal with times spent in different glucose ranges in one day. This work proposes a CoDa approach applied to glucose profiles obtained from six T1D patients using continuous glucose monitor (CGM). Glucose profiles of 24-h and 6-h duration were categorized according to the relative interpretation of time spent in different glucose ranges, with the objective of presenting a probabilistic model of prediction of category of the next 6-h period based on the category of the previous 24-h period. A discriminant model for determining the category of the 24-h periods was obtained, achieving an average above 94% of correct classification. A probabilistic model of transition between the category of the past 24-h of glucose to the category of the future 6-h period was obtained. Results show that the approach based on CoDa is suitable for the categorization of glucose profiles giving rise to a new analysis tool. This tool could be very helpful for patients, to anticipate the occurrence of potential adverse events or undesirable variability and for physicians to assess patients' outcomes and then tailor their therapies.
Collapse
|
43
|
Sepasi S, Kalat AA, Seyedabadi M. An adaptive back-stepping control for blood glucose regulation in type 1 diabetes. Biomed Signal Process Control 2021. [DOI: 10.1016/j.bspc.2021.102498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
44
|
Babar SA, Ahmad I, Mughal IS. Sliding-mode-based controllers for automation of blood glucose concentration for type 1 diabetes. IET Syst Biol 2021; 15:72-82. [PMID: 33780148 PMCID: PMC8675841 DOI: 10.1049/syb2.12015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 12/07/2020] [Accepted: 01/20/2021] [Indexed: 12/05/2022] Open
Abstract
Destruction of β-cells in pancreas causes deficiency in insulin production that leads to diabetes in the human body. To cope with this problem, insulin is either taken orally during the day or injected into the patient's body using artificial pancreas (AP) during sleeping hours. Some mathematical models indicate that AP uses control algorithms to regulate blood glucose concentration (BGC). The extended Bergman minimal model (EBMM) incorporates, as a state variable, the disturbance in insulin level during medication due to either meal intake or burning sugar by engaging in physical exercise. In this research work, EBMM and proposed finite time robust controllers are used, including the sliding mode controller (SMC), backstepping SMC (BSMC) and supertwisting SMC (second-order SMC or SOSMC) for automatic stabilisation of BGC in type 1 diabetic patients. The proposed SOSMC diminishes the chattering phenomenon which appears in the conventional SMC. The proposed BSMC is a recursive technique which becomes robust by the addition of the SMC. Lyapunov theory has been used to prove the asymptotic stability of the proposed controllers. Simulations have been carried out in MATLAB/Simulink for the comparative study of the proposed controllers under varying data of six different type 1 diabetic patients available in the literature.
Collapse
Affiliation(s)
- Sheraz Ahmad Babar
- School of Electrical Engineering and Computer Science (SEECS)National University of Sciences and Technology (NUST)IslamabadPakistan
| | - Iftikhar Ahmad
- School of Electrical Engineering and Computer Science (SEECS)National University of Sciences and Technology (NUST)IslamabadPakistan
| | - Iqra Shafeeq Mughal
- School of Electrical Engineering and Computer Science (SEECS)National University of Sciences and Technology (NUST)IslamabadPakistan
| |
Collapse
|
45
|
Millard LAC, Patel N, Tilling K, Lewcock M, Flach PA, Lawlor DA. GLU: a software package for analysing continuously measured glucose levels in epidemiology. Int J Epidemiol 2021; 49:744-757. [PMID: 32737505 PMCID: PMC7394960 DOI: 10.1093/ije/dyaa004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 01/09/2020] [Indexed: 12/22/2022] Open
Abstract
Continuous glucose monitors (CGM) record interstitial glucose levels 'continuously', producing a sequence of measurements for each participant (e.g. the average glucose level every 5 min over several days, both day and night). To analyse these data, researchers tend to derive summary variables such as the area under the curve (AUC), to then use in subsequent analyses. To date, a lack of consistency and transparency of precise definitions used for these summary variables has hindered interpretation, replication and comparison of results across studies. We present GLU, an open-source software package for deriving a consistent set of summary variables from CGM data. GLU performs quality control of each CGM sample (e.g. addressing missing data), derives a diverse set of summary variables (e.g. AUC and proportion of time spent in hypo-, normo- and hyper- glycaemic levels) covering six broad domains, and outputs these (with quality control information) to the user. GLU is implemented in R and is available on GitHub at https://github.com/MRCIEU/GLU. Git tag v0.2 corresponds to the version presented here.
Collapse
Affiliation(s)
- Louise A C Millard
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK.,Intelligent Systems Laboratory, Department of Computer Science, University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Nashita Patel
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, UK
| | - Kate Tilling
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Melanie Lewcock
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Peter A Flach
- Intelligent Systems Laboratory, Department of Computer Science, University of Bristol, Bristol, UK
| | - Debbie A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.,Bristol NIHR Biomedical Research Centre, Bristol, UK
| |
Collapse
|
46
|
Kdekian A, Sietzema M, Scherjon SA, Lutgers H, van der Beek EM. Pregnancy Outcomes and Maternal Insulin Sensitivity: Design and Rationale of a Multi-Center Longitudinal Study in Mother and Offspring (PROMIS). J Clin Med 2021; 10:jcm10050976. [PMID: 33801180 PMCID: PMC7957868 DOI: 10.3390/jcm10050976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 01/01/2023] Open
Abstract
The worldwide prevalence of overweight and obesity in women of reproductive age is rapidly increasing and a risk factor for the development of gestational diabetes (GDM). Excess adipose tissue reduces insulin sensitivity and may underlie adverse outcomes in both mother and child. The present paper describes the rationale and design of the PRegnancy Outcomes and Maternal Insulin Sensitivity (PROMIS) study, an exploratory cohort study to obtain detailed insights in insulin sensitivity and glucose metabolism during pregnancy and its relation to pregnancy outcomes including early infancy growth. We aim to recruit healthy pregnant women with a body mass index (BMI) ≥ 25 kg/m2 before 12 weeks of gestation in Northern Netherlands. A total of 130 woman will be checked on fasted (≤7.0 mmol/L) or random (≤11.0 mmol/L) blood glucose to exclude pregestational diabetes at inclusion. Subjects will be followed up to six months after giving birth, with a total of nine contact moments for data collection. Maternal data include postprandial measures following an oral meal tolerance test (MTT), conducted before 16 weeks and repeated around 24 weeks of gestation, followed by a standard oral glucose tolerance test before 28 weeks of gestation. The MTT is again performed around three months postpartum. Blood analysis is done for baseline and postprandial glucose and insulin, baseline lipid profile and several biomarkers of placental function. In addition, specific body circumferences, skinfold measures, and questionnaires about food intake, eating behavior, physical activity, meal test preference, mental health, and pregnancy complications will be obtained. Fetal data include assessment of growth, examined by sonography at week 28 and 32 of gestation. Neonatal and infant data consist of specific body circumferences, skinfolds, and body composition measurements, as well as questionnaires about eating behavior and complications up to 6 months after birth. The design of the PROMIS study will allow for detailed insights in the metabolic changes in the mother and their possible association with fetal and postnatal infant growth and body composition. We anticipate that the data from this cohort women with an elevated risk for the development of GDM may provide new insights to detect metabolic deviations already in early pregnancy. These data could inspire the development of new interventions that may improve the management of maternal, as well as offsrping complications from already early on in pregnancy with the aim to prevent adverse outcomes for mother and child.
Collapse
Affiliation(s)
- Anoush Kdekian
- Laboratory of Pediatrics, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
| | - Maaike Sietzema
- Clinical Endocrinology, Medical Centre Leeuwarden, Henri Dunantweg 2, 8934 AD Leeuwarden, The Netherlands; (M.S.); (H.L.)
| | - Sicco A. Scherjon
- Department of Gynaecology and Obstectrics, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
| | - Helen Lutgers
- Clinical Endocrinology, Medical Centre Leeuwarden, Henri Dunantweg 2, 8934 AD Leeuwarden, The Netherlands; (M.S.); (H.L.)
| | - Eline M. van der Beek
- Laboratory of Pediatrics, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
- Correspondence:
| |
Collapse
|
47
|
Pinnaro C, Christensen GE, Curtis V. Modeling Ketogenesis for Use in Pediatric Diabetes Simulation. J Diabetes Sci Technol 2021; 15:303-308. [PMID: 31608650 PMCID: PMC8256079 DOI: 10.1177/1932296819882058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Simulation is being increasingly integrated into medical education. Diabetes simulation is well-received by trainees and has demonstrated improved clinical results, including reduced adult inpatient hyperglycemia. However, no pediatric-specific diabetes simulation programs exist for use in medical education. None of the existing diabetes models incorporate ketones as an input or an output, which is essential for use in teaching pediatric diabetes management. METHODS We created a pediatric diabetes simulation incorporating both blood sugar and urine ketones as output. Ketone output is implemented as a state variable but is obfuscated to simulate hospital experience. Blood sugar output is similar to other models and incorporates the current blood sugar, insulin on board (IOB) and carbohydrates on board (COB), and insulin and carbohydrate sensitivities. The program calculates all IOB and COB every 15 minutes based on user input and provides written summary feedback at the end of the simulation about inaccurate dosing and timing. RESULTS The simulation realistically incorporated both blood glucose and urine ketones in clinically valid and actionable formats. After completing this simulation, 16/17 pediatric residents indicated that they wanted more simulated diabetes cases integrated into their curriculum. CONCLUSION Implementing simulation into pediatric diabetes education was feasible and well-received. More work is needed to further study the role of simulation in pediatric diabetes education when used adjunctively or in lieu of lectures when time or resources are limited.
Collapse
Affiliation(s)
- Catherina Pinnaro
- Stead Family Department of
Pediatrics, Department of Endocrinology and Diabetes, University of Iowa,
IA, USA
- Catherina Pinnaro, MD, Stead Family
Department of Pediatrics, Department of Endocrinology and Diabetes,
University of Iowa, 2015-20 Boyd Tower, 200 Hawkins Drive, Iowa City,
IA 52242, USA.
| | - Gary E. Christensen
- Department of Electrical and
Computer Engineering, University of Iowa, IA, USA
- Department of Radiation Oncology,
University of Iowa, IA, USA
| | - Vanessa Curtis
- Stead Family Department of
Pediatrics, Department of Endocrinology and Diabetes, University of Iowa,
IA, USA
| |
Collapse
|
48
|
Smaoui MR, Rabasa-Lhoret R, Haidar A. Development platform for artificial pancreas algorithms. PLoS One 2020; 15:e0243139. [PMID: 33332411 PMCID: PMC7746189 DOI: 10.1371/journal.pone.0243139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/17/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND AIMS Assessing algorithms of artificial pancreas systems is critical in developing automated and fault-tolerant solutions that work outside clinical settings. The development and evaluation of algorithms can be facilitated with a platform that conducts virtual clinical trials. We present in this paper a clinically validated cloud-based distributed platform that supports the development and comprehensive testing of single and dual-hormone algorithms for type 1 diabetes mellitus (T1DM). METHODS The platform is built on principles of object-oriented design and runs user algorithms in real-time virtual clinical trials utilizing a multi-threaded environment enabled by concurrent execution over a cloud infrastructure. The platform architecture isolates user algorithms located on personal machines from proprietary patient data running on the cloud. Users import a plugin into their algorithms (Matlab, Python, or Java) to connect to the platform. Once connected, users interact with a graphical interface to design experimental protocols for their trials. Protocols include trial duration in days, mealtimes and amounts, variability in mealtimes and amounts, carbohydrate counting errors, snacks, and onboard insulin levels. RESULTS The platform facilitates development by solving the ODE model in the cloud on large CPU-optimized machines, providing a 62% improvement in memory, speed and CPU utilization. Users can easily debug & modify code, test multiple strategies, and generate detailed clinical performance reports. We validated and integrated into the platform a glucoregulatory system of ordinary differential equations (ODEs) parameterized with clinical data to mimic the inter and intra-day variability of glucose responses of 15 T1DM patients. CONCLUSION The platform utilizes the validated patient model to conduct virtual clinical trials for the rapid development and testing of closed-loop algorithms for T1DM.
Collapse
Affiliation(s)
- Mohamed Raef Smaoui
- Computer Science Department, Faculty of Science, Kuwait University, Kuwait City, Kuwait
- * E-mail:
| | - Remi Rabasa-Lhoret
- Department of Nutrition, Faculty of Medicine, Université de Montréal, Montréal, Canada
- Institut de Recherches Cliniques de Montréal, Montréal, Canada
| | - Ahmad Haidar
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Canada
| |
Collapse
|
49
|
Alam W, Khan Q, Riaz RA, Akmeliawati R. Arbitrary-order sliding mode-based robust control algorithm for the developing artificial pancreas mechanism. IET Syst Biol 2020; 14:307-313. [PMID: 33399094 PMCID: PMC8687268 DOI: 10.1049/iet-syb.2018.5075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 04/15/2020] [Accepted: 05/27/2020] [Indexed: 11/20/2022] Open
Abstract
In Diabetes Mellitus, the pancreas remains incapable of insulin administration that leads to hyperglycaemia, an escalated glycaemic concentration, which may stimulate many complications. To circumvent this situation, a closed-loop control strategy is much needed for the exogenous insulin infusion in diabetic patients. This closed-loop structure is often termed as an artificial pancreas that is generally established by the employment of different feedback control strategies. In this work, the authors have proposed an arbitrary-order sliding mode control approach for development of the said mechanism. The term, arbitrary, is exercised in the sense of its applicability to any n-order controllable canonical system. The proposed control algorithm affirms the finite-time effective stabilisation of the glucose-insulin regulatory system, at the desired level, with the alleviation of sharp fluctuations. The novelty of this work lies in the sliding manifold that incorporates indirect non-linear terms. In addition, the necessary discontinuous terms are filtered-out once before its employment to the plant, i.e. diabetic patient. The robustness, in the presence of external disturbances, i.e. meal intake is confirmed via rigorous mathematical stability analysis. In addition, the effectiveness of the proposed control strategy is ascertained by comparing the results with the standard literature.
Collapse
Affiliation(s)
- Waqar Alam
- Department of Electrical and Computer Engineering, COMSATS University Islamabad, Islamabad, Pakistan
| | - Qudrat Khan
- Center for Advanced Studies in Telecommunications (CAST), COMSATS University Islamabad, Islamabad, Pakistan.
| | - Raja Ali Riaz
- Department of Electrical and Computer Engineering, COMSATS University Islamabad, Islamabad, Pakistan
| | - Rini Akmeliawati
- School of Mechanical Engineering, The University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
50
|
Batmani Y, Khodakaramzadeh S. Blood glucose concentration control for type 1 diabetic patients: a multiple-model strategy. IET Syst Biol 2020; 14:24-30. [PMID: 31931478 DOI: 10.1049/iet-syb.2018.5049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, a multiple-model strategy is evaluated as an alternative closed-loop method for subcutaneous insulin delivery in type 1 diabetes. Non-linearities of the glucose-insulin regulatory system are considered by modelling the system around five different operating points. After conducting some identification experiments in the UVA/Padova metabolic simulator (accepted simulator by the US Food and Drug Administration (FDA)), five transfer functions are obtained for these operating points. Paying attention to some physiological facts, the control objectives such as the required settling time and permissible bounds of overshoots and undershoots are determined for any transfer functions. Then, five PID controllers are tuned to achieve these objectives and a bank of controllers is constructed. To cope with difficulties of the presence of delays in subcutaneous blood glucose (BG) measuring and in administration of insulin, a glucose-dependent setpoint is considered as the desired trajectory for the BG concentration. The performance of the obtained closed-loop glucose-insulin regulatory system is investigated on the in silico adult cohort of the UVA/Padova metabolic simulator. The obtained results show that the proposed multiple-model strategy leads to a closed-loop mechanism with limited hyperglycemia and no severe hypoglycemia.
Collapse
Affiliation(s)
- Yazdan Batmani
- Department of Electrical Engineering, University of Kurdistan, Sanandaj, Iran.
| | | |
Collapse
|