1
|
Feng S, Zhang Y, Hou C, Liu Y, Gao Y, Song Y, Luo J. A temperature-responsive dual-hormone foam nanoengine improves rectal absorptivity of insulin-pramlintide for diabetes treatment. SCIENCE ADVANCES 2024; 10:eadn8695. [PMID: 39196940 PMCID: PMC11352908 DOI: 10.1126/sciadv.adn8695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 07/24/2024] [Indexed: 08/30/2024]
Abstract
Despite the therapeutic benefits of insulin-pramlintide dual-hormone therapy in diabetes, its application potential has been limited due to a lack of efficient delivery routes. Here, we developed a temperature-responsive dual-hormone foam nanoengine (HormFoam) and combined it with a customized spraying device to further construct an in situ foam-generating system for improving the rectal bioavailability of dual-hormone therapy. To support rapid clinical translation, a continuous microfluidic preparation for HormFoam was proposed, including the power unit of perfluorocarbon nanodroplets and the pharmaceutical components Pluronic F127-functionalized liposomal insulin and pramlintide. We found that HormFoam could consistently generate foams to drive drugs forward after rectal administration, which enhanced intestinal distribution and mucosa absorption, leading to systemic codelivery of insulin-pramlintide. HormFoam reproduced the physiology of endocrine pancreas for glycemic control and induced body weight loss while reversing metabolic disorders in diabetic mice with good biosafety. Therefore, HormFoam represents a state-of-the-art dual-hormone regimen with the potential to address unmet needs in diabetes management.
Collapse
Affiliation(s)
- Shujun Feng
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
- College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| | - Yu Zhang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Chunyuan Hou
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Yuta Liu
- College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| | - Yanfeng Gao
- College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
- School of Medical Imaging, Wannan Medical College, Wuhu 241002, China
| | - Yujun Song
- College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| | - Jun Luo
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| |
Collapse
|
2
|
Furió-Novejarque C, Sala-Mira I, Díez JL, Bondia J. A model of subcutaneous pramlintide pharmacokinetics and its effect on gastric emptying: Proof-of-concept based on populational data. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 244:107968. [PMID: 38064957 DOI: 10.1016/j.cmpb.2023.107968] [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: 09/18/2023] [Revised: 11/12/2023] [Accepted: 12/01/2023] [Indexed: 01/26/2024]
Abstract
Pramlintide, an amylin analog, has been coming up as an agent in type 1 diabetes dual-hormone therapies (insulin/pramlintide). Since pramlintide slows down gastric emptying, it allows for easing glucose control and reducing the burden of meal announcements. Pre-clinical in silico evaluations are a key step in the development of any closed-loop strategy. However, mathematical models are needed, and pramlintide models in the literature are scarce. This work proposes a proof-of-concept pramlintide model, describing its subcutaneous pharmacokinetics (PK) and its effect on gastric emptying (PD). The model is validated with published populational (clinical) data. The model development is divided into three stages: intravenous PK, subcutaneous PK, and PD modeling. In each stage, a set of model structures are proposed, and their performance is assessed using the Akaike Information Criterion (AIC) and the Bayesian Information Criterion (BIC). In order to evaluate the modulation of the rate of gastric emptying, a literature meal model was used. The final pramlintide model comprises four compartments and a function that modulates gastric emptying depending on plasma pramlintide. Results show an appropriate fit for the data. Some aspects are left as open questions due to the lack of specific data (e.g., the influence of meal composition on the pramlintide effect). Moreover, further validation with individual data is necessary to propose a virtual cohort of patients.
Collapse
Affiliation(s)
- Clara Furió-Novejarque
- Instituto Universitario de Automática e Informática Industrial, Universitat Politècnica de València, C/ Camí de Vera, s/n, València, 46022, Spain.
| | - Iván Sala-Mira
- Instituto Universitario de Automática e Informática Industrial, Universitat Politècnica de València, C/ Camí de Vera, s/n, València, 46022, Spain.
| | - José-Luis Díez
- Instituto Universitario de Automática e Informática Industrial, Universitat Politècnica de València, C/ Camí de Vera, s/n, València, 46022, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5. Pabellón 11, Madrid, 28029, Spain.
| | - Jorge Bondia
- Instituto Universitario de Automática e Informática Industrial, Universitat Politècnica de València, C/ Camí de Vera, s/n, València, 46022, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5. Pabellón 11, Madrid, 28029, Spain.
| |
Collapse
|
3
|
Eržen S, Tonin G, Jurišić Eržen D, Klen J. Amylin, Another Important Neuroendocrine Hormone for the Treatment of Diabesity. Int J Mol Sci 2024; 25:1517. [PMID: 38338796 PMCID: PMC10855385 DOI: 10.3390/ijms25031517] [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: 12/30/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Diabetes mellitus is a devastating chronic metabolic disease. Since the majority of type 2 diabetes mellitus patients are overweight or obese, a novel term-diabesity-has emerged. The gut-brain axis plays a critical function in maintaining glucose and energy homeostasis and involves a variety of peptides. Amylin is a neuroendocrine anorexigenic polypeptide hormone, which is co-secreted with insulin from β-cells of the pancreas in response to food consumption. Aside from its effect on glucose homeostasis, amylin inhibits homeostatic and hedonic feeding, induces satiety, and decreases body weight. In this narrative review, we summarized the current evidence and ongoing studies on the mechanism of action, clinical pharmacology, and applications of amylin and its analogs, pramlintide and cagrilintide, in the field of diabetology, endocrinology, and metabolism disorders, such as obesity.
Collapse
Affiliation(s)
- Stjepan Eržen
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Gašper Tonin
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Arts, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Dubravka Jurišić Eržen
- Department of Endocrinology and Diabetology, University Hospital Centre, 51000 Rijeka, Croatia
- Department of Internal Medicine, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Jasna Klen
- Division of Surgery, Department of Abdominal Surgery, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
4
|
Almurashi AM, Rodriguez E, Garg SK. Emerging Diabetes Technologies: Continuous Glucose Monitors/Artificial Pancreases. J Indian Inst Sci 2023; 103:1-26. [PMID: 37362851 PMCID: PMC10043869 DOI: 10.1007/s41745-022-00348-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/04/2022] [Indexed: 03/30/2023]
Abstract
Over the past decade there have been many advances in diabetes technologies, such as continuous glucose monitors (CGM s), insulin-delivery devices, and hybrid closed loop systems . Now most CGMs (Medtronic-Guardian, Dexcom-G6, and Abbott-Libre-2) have MARD values of < 10%, in contrast to two decades ago when the MARD used to be > 20%. In addition, the majority of the new CGMs do not require calibrations, and the latest CGMs last for 10-14 days. An implantable 6-months CGM by Eversense-3 is now approved in the USA and Europe. Recently, the FDA approved Libre 3 which provides real-time glucose values every minute. Even though it is approved as an iCGM it is not interoperable with automatic-insulin-delivery (AID) systems. The newer CGMs that are likely to be launched in the next few months in the USA include the 10-11 days Dexcom G7 (60% smaller than the existing G6), and the 7-days Medtronic Guardian 4. Most of the newer CGM have several features like automatic initialization, easy insertion, predictive alarms, and alerts. It has also been noticed that an arm insertion site might have better accuracy than abdomen or other sites, like the buttock for kids. Lag time between YSI and different sensors have been reported differently, sometimes it is down to 2-3 min; however, in many instances, it is still 15-20 min, especially when the rate of change of glucose is > 2 mg/min. We believe that in the next decade there will be a significant increase in the number of people who use CGM for their day-to-day diabetes care.
Collapse
Affiliation(s)
- Abdulhalim M. Almurashi
- Barbara Davis Center for Diabetes, University of Colorado Denver, 1775 Aurora Ct, Rm 1324, Aurora, CO 80045 USA
- Madinah Health Cluster, Madinah, Saudi Arabia
| | - Erika Rodriguez
- Barbara Davis Center for Diabetes, University of Colorado Denver, 1775 Aurora Ct, Rm 1324, Aurora, CO 80045 USA
| | - Satish K. Garg
- Barbara Davis Center for Diabetes, University of Colorado Denver, 1775 Aurora Ct, Rm 1324, Aurora, CO 80045 USA
| |
Collapse
|
5
|
100 Years of insulin: A chemical engineering perspective. KOREAN J CHEM ENG 2023. [DOI: 10.1007/s11814-022-1308-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
6
|
Rodríguez-Sarmiento DL, León-Vargas F, García-Jaramillo M. Artificial pancreas systems: experiences from concept to commercialisation. Expert Rev Med Devices 2022; 19:877-894. [DOI: 10.1080/17434440.2022.2150546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
7
|
McCarthy O, Schmidt S, Christensen MB, Bain SC, Nørgaard K, Bracken R. The endocrine pancreas during exercise in people with and without type 1 diabetes: Beyond the beta-cell. Front Endocrinol (Lausanne) 2022; 13:981723. [PMID: 36147573 PMCID: PMC9485437 DOI: 10.3389/fendo.2022.981723] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Although important for digestion and metabolism in repose, the healthy endocrine pancreas also plays a key role in facilitating energy transduction around physical exercise. During exercise, decrements in pancreatic β-cell mediated insulin release opposed by increments in α-cell glucagon secretion stand chief among the hierarchy of glucose-counterregulatory responses to decreasing plasma glucose levels. As a control hub for several major glucose regulatory hormones, the endogenous pancreas is therefore essential in ensuring glucose homeostasis. Type 1 diabetes (T1D) is pathophysiological condition characterised by a destruction of pancreatic β-cells resulting in pronounced aberrations in glucose control. Yet beyond the beta-cell perhaps less considered is the impact of T1D on all other pancreatic endocrine cell responses during exercise and whether they differ to those observed in healthy man. For physicians, understanding how the endocrine pancreas responds to exercise in people with and without T1D may serve as a useful model from which to identify whether there are clinically relevant adaptations that need consideration for glycaemic management. From a physiological perspective, delineating differences or indeed similarities in such responses may help inform appropriate exercise test interpretation and subsequent program prescription. With more complex advances in automated insulin delivery (AID) systems and emerging data on exercise algorithms, a timely update is warranted in our understanding of the endogenous endocrine pancreatic responses to physical exercise in people with and without T1D. By placing our focus here, we may be able to offer a nexus of better understanding between the clinical and engineering importance of AIDs requirements during physical exercise.
Collapse
Affiliation(s)
- Olivia McCarthy
- Applied Sport, Technology, Exercise and Medicine Research Centre, Swansea University, Swansea, United Kingdom
- Steno Diabetes Center Copenhagen, Copenhagen University Hospital, Herlev, Denmark
- *Correspondence: Olivia McCarthy,
| | - Signe Schmidt
- Steno Diabetes Center Copenhagen, Copenhagen University Hospital, Herlev, Denmark
| | | | | | - Kirsten Nørgaard
- Steno Diabetes Center Copenhagen, Copenhagen University Hospital, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Richard Bracken
- Applied Sport, Technology, Exercise and Medicine Research Centre, Swansea University, Swansea, United Kingdom
| |
Collapse
|
8
|
Abstract
Closed-loop insulin delivery systems are fast becoming the standard of care in the management of type 1 diabetes and have led to significant improvements in diabetes management. Nevertheless, there is still room for improvement for the closed-loop systems to optimize treatment and meet target glycemic control. Adjunct treatments have been introduced as an alternative method to insulin-only treatment methods to overcome diabetes treatment challenges and improve clinical and patient reported outcomes during closed-loop treatment. The adjunct treatment agents mostly consist of medications that are already approved for type 2 diabetes treatment and aim to complete the missing physiologic factors, such as the entero-endocrine system, that regulate glycemia in addition to insulin. This paper will review many of these adjunct therapies, including the basic mechanisms of action, potential benefits, side effects, and the evidence supporting their use during closed-loop treatment.
Collapse
Affiliation(s)
- Shylaja Srinivasan
- Division of Pediatric Endocrinology and
Diabetes, University of San Francisco, CA, USA
| | - Laya Ekhlaspour
- Division of Pediatric Endocrinology and
Diabetes, Stanford University, Palo Alto, CA, USA
| | - Eda Cengiz
- Division of Pediatric Endocrinology and
Diabetes, Yale University, New Haven, NJ, USA
| |
Collapse
|
9
|
Maikawa CL, Chen PC, Vuong ET, Nguyen LT, Mann JL, d'Aquino AI, Lal RA, Maahs DM, Buckingham BA, Appel EA. Ultra-Fast Insulin-Pramlintide Co-Formulation for Improved Glucose Management in Diabetic Rats. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101575. [PMID: 34499434 PMCID: PMC8564421 DOI: 10.1002/advs.202101575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/07/2021] [Indexed: 05/04/2023]
Abstract
Dual-hormone replacement therapy with insulin and amylin in patients with type 1 diabetes has the potential to improve glucose management. Unfortunately, currently available formulations require burdensome separate injections at mealtimes and have disparate pharmacokinetics that do not mimic endogenous co-secretion. Here, amphiphilic acrylamide copolymers are used to create a stable co-formulation of monomeric insulin and amylin analogues (lispro and pramlintide) with synchronous pharmacokinetics and ultra-rapid action. The co-formulation is stable for over 16 h under stressed aging conditions, whereas commercial insulin lispro (Humalog) aggregates in 8 h. The faster pharmacokinetics of monomeric insulin in this co-formulation result in increased insulin-pramlintide overlap of 75 ± 6% compared to only 47 ± 7% for separate injections. The co-formulation results in similar delay in gastric emptying compared to pramlintide delivered separately. In a glucose challenge, in rats, the co-formulation reduces deviation from baseline glucose compared to insulin only, or separate insulin and pramlintide administrations. Further, comparison of interspecies pharmacokinetics of monomeric pramlintide suggests that pharmacokinetics observed for the co-formulation will be well preserved in future translation to humans. Together these results suggest that the co-formulation has the potential to improve mealtime glucose management and reduce patient burden in the treatment of diabetes.
Collapse
Affiliation(s)
- Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Peyton C Chen
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Eric T Vuong
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Leslee T Nguyen
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Joseph L Mann
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Andrea I d'Aquino
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Rayhan A Lal
- Department of Medicine (Endocrinology), Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA, 94305, USA
| | - David M Maahs
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA, 94305, USA
| | - Bruce A Buckingham
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA, 94305, USA
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, 94305, USA
- Diabetes Research Center, Stanford University, Stanford, CA, 94305, USA
- ChEM-H Institute, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
10
|
Pasqua MR, Tsoukas MA, Haidar A. Strategically Playing with Fire: SGLT Inhibitors as Possible Adjunct to Closed-Loop Insulin Therapy. J Diabetes Sci Technol 2021; 15:1232-1242. [PMID: 34558336 PMCID: PMC8655283 DOI: 10.1177/19322968211035411] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As closed-loop insulin therapies emerge into clinical practice and evolve in medical research for type 1 diabetes (T1D) treatment, the limitations in these therapies become more evident. These gaps include unachieved target levels of glycated hemoglobin in some patients, postprandial hyperglycemia, the ongoing need for carbohydrate counting, and the lack of non-glycemic benefits (such as prevention of metabolic syndrome and complications). Multiple adjunct therapies have been examined to improve closed-loop systems, yet none have become a staple. Sodium-glucose-linked cotransporter inhibitors (SGLTi's) have been extensively researched in T1D, with average reductions in placebo-adjusted HbA1c by 0.39%, and total daily dose by approximately 10%. Unfortunately, many trials revealed an increased risk of diabetic ketoacidosis, as high as 5 times the relative risk compared to placebo. This narrative review discusses the proven benefits and risks of SGLTi in patients with T1D with routine therapy, what has been studied thus far in closed-loop therapy in combination with SGLTi, the potential benefits of SGLTi use to closed-loop systems, and what is required going forward to improve the benefit to risk ratio in these insulin systems.
Collapse
Affiliation(s)
- Melissa-Rosina Pasqua
- Division of Endocrinology, McGill
University Health Centre, Montreal, QC, Canada
- Melissa-Rosina Pasqua, MD, Mailing address:
McGill University Health Centre, 1001 boulevard de Décarie, Montreal, QC H4A
3J1, Canada.
| | - Michael A. Tsoukas
- Division of Endocrinology, McGill
University Health Centre, Montreal, QC, Canada
| | - Ahmad Haidar
- Department of Biomedical Engineering,
McGill University, Montreal, QC, Canada
| |
Collapse
|
11
|
Moon SJ, Jung I, Park CY. Current Advances of Artificial Pancreas Systems: A Comprehensive Review of the Clinical Evidence. Diabetes Metab J 2021; 45:813-839. [PMID: 34847641 PMCID: PMC8640161 DOI: 10.4093/dmj.2021.0177] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/24/2021] [Indexed: 12/19/2022] Open
Abstract
Since Banting and Best isolated insulin in the 1920s, dramatic progress has been made in the treatment of type 1 diabetes mellitus (T1DM). However, dose titration and timely injection to maintain optimal glycemic control are often challenging for T1DM patients and their families because they require frequent blood glucose checks. In recent years, technological advances in insulin pumps and continuous glucose monitoring systems have created paradigm shifts in T1DM care that are being extended to develop artificial pancreas systems (APSs). Numerous studies that demonstrate the superiority of glycemic control offered by APSs over those offered by conventional treatment are still being published, and rapid commercialization and use in actual practice have already begun. Given this rapid development, keeping up with the latest knowledge in an organized way is confusing for both patients and medical staff. Herein, we explore the history, clinical evidence, and current state of APSs, focusing on various development groups and the commercialization status. We also discuss APS development in groups outside the usual T1DM patients and the administration of adjunct agents, such as amylin analogues, in APSs.
Collapse
Affiliation(s)
- Sun Joon Moon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Inha Jung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Cheol-Young Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
12
|
Beneyto A, Bequette BW, Vehi J. Fault Tolerant Strategies for Automated Insulin Delivery Considering the Human Component: Current and Future Perspectives. J Diabetes Sci Technol 2021; 15:1224-1231. [PMID: 34286613 PMCID: PMC8655284 DOI: 10.1177/19322968211029297] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Automated Insulin Delivery (AID) are systems developed for daily use by people with type 1 diabetes (T1D). To ensure the safety of users, it is essential to consider how the human factor affects the performance and safety of these devices. While there are numerous publications on hardware-related failures of AID systems, there are few studies on the human component of the system. From a control point of view, people with T1D using AID systems are at the same time the plant to be controlled and the plant operator. Therefore, users may induce faults in the controller, sensors, actuators, and the plant itself. Strategies to cope with the human interaction in AID systems are needed for further development of the technology. In this paper, we present an analysis of potential faults introduced by AID users when the system is under normal operation. This is followed by a review of current fault tolerant control (FTC) approaches to identify missing areas of research. The paper concludes with a discussion on future directions for the new generation of FTC AID systems.
Collapse
Affiliation(s)
| | | | - Josep Vehi
- Universitat de Girona, Girona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| |
Collapse
|
13
|
Ciechanowska A, Gora I, Sabalinska S, Foltynski P, Ladyzynski P. Effect of glucose concentration and culture substrate on HUVECs viability in in vitro cultures: A literature review and own results. Biocybern Biomed Eng 2021. [DOI: 10.1016/j.bbe.2021.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
14
|
Tsoukas MA, Majdpour D, Yale JF, Fathi AE, Garfield N, Rutkowski J, Rene J, Legault L, Haidar A. A fully artificial pancreas versus a hybrid artificial pancreas for type 1 diabetes: a single-centre, open-label, randomised controlled, crossover, non-inferiority trial. LANCET DIGITAL HEALTH 2021; 3:e723-e732. [PMID: 34580055 DOI: 10.1016/s2589-7500(21)00139-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/14/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND For people with type 1 diabetes, there is currently no automated insulin delivery system that does not require meal input. We aimed to assess the efficacy of a novel faster-acting insulin aspart (Fiasp) plus pramlintide fully closed-loop system that does not require meal input. METHODS In this open-label, randomised controlled, crossover, non-inferiority trial we compared the Fiasp (Novo Nordisk, Bagsværd, Denmark) plus pramlintide closed-loop system with no meal input (fully artificial pancreas) and the Fiasp-alone closed-loop system with precise carbohydrate counting (hybrid artificial pancreas). Adults (≥18 years) who had a clinical diagnosis of type 1 diabetes for at least 12 months, had glycated haemoglobin 12% or lower, and had been on insulin pump therapy for at least 6 months were enrolled at McGill University Health Centre, Montreal, QC, Canada. The Fiasp plus pramlintide fully closed-loop system delivered pramlintide in a basal-bolus manner with a fixed ratio of 10 μg:U relative to insulin. A research staff member counted the carbohydrate content of meals to input in the hybrid closed-loop system. Participants completed the two full-day crossover interventions in a random order allocated by a computer-generated code implementing a blocked randomisation (block size of four). The primary outcome was the percentage of time spent within the glucose target range (3·9-10·0 mmol/L), with a 6% non-inferiority margin, assessed in all participants who completed both interventions. This trial is registered with ClinicalTrials.gov, NCT03800875. FINDINGS Between Feb 8, 2019, and Sept 19, 2020, we enrolled 28 adults, of whom 24 completed both interventions and were included in analyses. The percentage of time spent in the target range was 74·3% (IQR 61·5-82·8) with the fully closed-loop system versus 78·1% (66·3-87·5) with the hybrid Fiasp-alone closed-loop system (paired difference 2·6%, 95% CI -2·4 to 12·2; non-inferiority p=0·28). Eight (33%) participants had at least one hypoglycaemia event (<3·3 mmol/L) with the fully closed-loop system compared with 14 (58%) participants with the hybrid closed-loop system (2200-2200 h). Non-mild nausea was reported by three (13%) participants and non-mild bloating by one (4%) participant with the fully closed-loop system compared with zero participants with the hybrid closed-loop system. INTERPRETATION The Fiasp plus pramlintide fully closed-loop system was not non-inferior to the Fiasp-alone hybrid closed-loop system for the overall percentage of time in the glucose target range. However, participants still spent a high percentage of time within the target range with the fully-closed loop system. Outpatient studies comparing the fully closed-loop hybrid systems with patient-estimated, rather than precise, carbohydrate counting are warranted. FUNDING Diabetes Canada.
Collapse
Affiliation(s)
- Michael A Tsoukas
- Division of Endocrinology and Metabolism, Royal Victoria Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Dorsa Majdpour
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Jean-François Yale
- Division of Endocrinology and Metabolism, Royal Victoria Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Anas El Fathi
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Natasha Garfield
- Division of Endocrinology and Metabolism, Royal Victoria Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Joanna Rutkowski
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Jennifer Rene
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Laurent Legault
- Division of Endocrinology and Metabolism, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Ahmad Haidar
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada.
| |
Collapse
|
15
|
Tsoukas MA, Cohen E, Legault L, von Oettingen JE, Yale JF, Vallis M, Odabassian M, El Fathi A, Rutkowski J, Jafar A, Ghanbari M, Gouchie-Provencher N, René J, Palisaitis E, Haidar A. Alleviating carbohydrate counting with a FiASP-plus-pramlintide closed-loop delivery system (artificial pancreas): Feasibility and pilot studies. Diabetes Obes Metab 2021; 23:2090-2098. [PMID: 34047449 DOI: 10.1111/dom.14447] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/18/2021] [Accepted: 05/23/2021] [Indexed: 01/07/2023]
Abstract
AIM To assess whether a FiASP-and-pramlintide closed-loop system has the potential to replace carbohydrate counting with a simple meal announcement (SMA) strategy (meal priming bolus without carbohydrate counting) without degrading glycaemic control compared with a FiASP closed-loop system. MATERIALS AND METHODS We conducted a 24-hour feasibility study comparing a FiASP system with full carbohydrate counting (FCC) with a FiASP-and-pramlintide system with SMA. We conducted a subsequent 12-day outpatient pilot study comparing a FiASP-and-placebo system with FCC, a FiASP-and-pramlintide system with SMA, and a FiASP-and-placebo system with SMA. Basal-bolus FiASP-and-pramlintide were delivered at a fixed ratio (1 U:10 μg). Glycaemic outcomes were measured, surveys evaluated gastrointestinal symptoms and diabetes distress, and participant interviews helped establish a preliminary coding framework to assess user experience. RESULTS Seven participants were included in the feasibility analysis. Time spent in 3.9-10 mmol/L was similar between both interventions (81%-84%). Four participants were included in the pilot analysis. Time spent in 3.9-10 mmol/L was similar between the FiASP-and-placebo with FCC and FiASP-and-pramlintide with SMA interventions (70%), but was lower in the FiASP-and-placebo with SMA intervention (60%). Time less than 3.9 mmol/L and gastrointestinal symptoms were similar across all interventions. Emotional distress was moderate at baseline, after the FiASP-and-placebo with FCC and SMA interventions, and fell after the FiASP-and-pramlintide with SMA intervention. SMA reportedly afforded participants flexibility and reduced mealtime concerns. CONCLUSIONS The FiASP-and-pramlintide system has the potential to substitute carbohydrate counting with SMA without degrading glucose control.
Collapse
Affiliation(s)
- Michael A Tsoukas
- Division of Endocrinology, McGill University Health Centre, Montreal, Canada
- The Research Institute of McGill University Health Centre, Montreal, Canada
| | - Elisa Cohen
- Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Laurent Legault
- The Research Institute of McGill University Health Centre, Montreal, Canada
- Department of Pediatrics, Division of Pediatric Endocrinology, McGill University Health Centre, Montreal, Canada
| | - Julia E von Oettingen
- The Research Institute of McGill University Health Centre, Montreal, Canada
- Department of Pediatrics, Division of Pediatric Endocrinology, McGill University Health Centre, Montreal, Canada
| | - Jean-François Yale
- Division of Endocrinology, McGill University Health Centre, Montreal, Canada
- The Research Institute of McGill University Health Centre, Montreal, Canada
| | - Michael Vallis
- Department of Family Medicine, Dalhousie University, Halifax, Canada
| | - Madison Odabassian
- Department of Biomedical Engineering, McGill University, Montreal, Canada
| | - Anas El Fathi
- Department of Biomedical Engineering, McGill University, Montreal, Canada
| | - Joanna Rutkowski
- Department of Biomedical Engineering, McGill University, Montreal, Canada
| | - Adnan Jafar
- Department of Biomedical Engineering, McGill University, Montreal, Canada
| | - Milad Ghanbari
- Department of Biomedical Engineering, McGill University, Montreal, Canada
| | | | - Jennifer René
- The Research Institute of McGill University Health Centre, Montreal, Canada
| | - Emilie Palisaitis
- Department of Biomedical Engineering, McGill University, Montreal, Canada
| | - Ahmad Haidar
- The Research Institute of McGill University Health Centre, Montreal, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Canada
| |
Collapse
|
16
|
Infante M, Baidal DA, Rickels MR, Fabbri A, Skyler JS, Alejandro R, Ricordi C. Dual-hormone artificial pancreas for management of type 1 diabetes: Recent progress and future directions. Artif Organs 2021; 45:968-986. [PMID: 34263961 PMCID: PMC9059950 DOI: 10.1111/aor.14023] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023]
Abstract
Over the last few years, technological advances have led to tremendous improvement in the management of type 1 diabetes (T1D). Artificial pancreas systems have been shown to improve glucose control compared with conventional insulin pump therapy. However, clinically significant hypoglycemic and hyperglycemic episodes still occur with the artificial pancreas. Postprandial glucose excursions and exercise-induced hypoglycemia represent major hurdles in improving glucose control and glucose variability in many patients with T1D. In this regard, dual-hormone artificial pancreas systems delivering other hormones in addition to insulin (glucagon or amylin) may better reproduce the physiology of the endocrine pancreas and have been suggested as an alternative tool to overcome these limitations in clinical practice. In addition, novel ultra-rapid-acting insulin analogs with a more physiological time-action profile are currently under investigation for use in artificial pancreas devices, aiming to address the unmet need for further improvements in postprandial glucose control. This review article aims to discuss the current progress and future outlook in the development of novel ultra-rapid insulin analogs and dual-hormone closed-loop systems, which offer the next steps to fully closing the loop in the artificial pancreas.
Collapse
Affiliation(s)
- Marco Infante
- Clinical Cell Transplant Program, Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Division of Endocrinology, Metabolism and Diabetes, Department of Systems Medicine, CTO A. Alesini Hospital, Diabetes Research Institute Federation, University of Rome Tor Vergata, Rome, Italy
- UniCamillus, Saint Camillus International University of Health Sciences, Rome, Italy
| | - David A. Baidal
- Clinical Cell Transplant Program, Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael R. Rickels
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Andrea Fabbri
- Division of Endocrinology, Metabolism and Diabetes, Department of Systems Medicine, CTO A. Alesini Hospital, Diabetes Research Institute Federation, University of Rome Tor Vergata, Rome, Italy
| | - Jay S. Skyler
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rodolfo Alejandro
- Clinical Cell Transplant Program, Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Camillo Ricordi
- Clinical Cell Transplant Program, Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
17
|
Shang T, Zhang JY, Bequette BW, Raymond JK, Coté G, Sherr JL, Castle J, Pickup J, Pavlovic Y, Espinoza J, Messer LH, Heise T, Mendez CE, Kim S, Ginsberg BH, Masharani U, Galindo RJ, Klonoff DC. Diabetes Technology Meeting 2020. J Diabetes Sci Technol 2021; 15:916-960. [PMID: 34196228 PMCID: PMC8258529 DOI: 10.1177/19322968211016480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [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
Diabetes Technology Society hosted its annual Diabetes Technology Meeting on November 12 to November 14, 2020. This meeting brought together speakers to cover various perspectives about the field of diabetes technology. The meeting topics included artificial intelligence, digital health, telemedicine, glucose monitoring, regulatory trends, metrics for expressing glycemia, pharmaceuticals, automated insulin delivery systems, novel insulins, metrics for diabetes monitoring, and discriminatory aspects of diabetes technology. A live demonstration was presented.
Collapse
Affiliation(s)
- Trisha Shang
- Diabetes Technology Society, Burlingame, CA, USA
| | | | | | - Jennifer K. Raymond
- Children’s Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Gerard Coté
- Texas A & M University, College Station, Texas, USA
| | | | | | | | | | - Juan Espinoza
- Children’s Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | | | | | | | - Sarah Kim
- University of California San Francisco, San Francisco, CA, USA
| | | | - Umesh Masharani
- University of California San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
18
|
Majdpour D, Tsoukas MA, Yale JF, El Fathi A, Rutkowski J, Rene J, Garfield N, Legault L, Haidar A. Fully Automated Artificial Pancreas for Adults With Type 1 Diabetes Using Multiple Hormones: Exploratory Experiments. Can J Diabetes 2021; 45:734-742. [PMID: 33888413 DOI: 10.1016/j.jcjd.2021.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 02/11/2021] [Accepted: 02/14/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVES A fully automated insulin-pramlintide-glucagon artificial pancreas that alleviates the burden of carbohydrate counting without degrading glycemic control was iteratively enhanced until convergence through pilot experiments on adults with type 1 diabetes. METHODS Nine participants (age, 37±13 years; glycated hemoglobin, 7.7±0.7%) completed two 27-hour interventions: a fully automated multihormone artificial pancreas and a comparator insulin-alone artificial pancreas with carbohydrate counting. The baseline algorithm was a model-predictive controller that administered insulin and pramlintide in a fixed ratio, with boluses triggered by a glucose threshold, and administered glucagon in response to low glucose levels. RESULTS The baseline multihormone dosing algorithm resulted in noninferior time in target range (3.9 to 10.0 mmol/L) (71%) compared with the insulin-alone arm (70%) in 2 participants, with minimal glucagon delivery. The algorithm was modified to deliver insulin and pramlintide more aggressively to increase time in range and maximize the benefits of glucagon. The modified algorithm displayed a similar time in range for the multihormone arm (79%) compared with the insulin-alone arm (83%) in 2 participants, but with undesired glycemic fluctuations. Subsequently, we reduced the glucose threshold that triggers glucagon boluses. This resulted in inferior glycemic control for the multihormone arm (81% vs 91%) in 2 participants. Thereafter, a model-based meal-detection algorithm to deliver insulin and pramlintide boluses closer to mealtimes was added and glucagon was removed. The final dual-hormone system had comparable time in range (81% vs 83%) in the last 3 participants. CONCLUSION The final version of the fully automated system that delivered insulin and pramlintide warrants a randomized controlled trial.
Collapse
Affiliation(s)
- Dorsa Majdpour
- Department of Biomedical Engineering, McGill University, Montréal, Québec, Canada; The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - Michael A Tsoukas
- The Research Institute of McGill University Health Centre, Montréal, Québec, Canada; Royal Victoria Hospital, McGill University Health Centre, Montréal, Québec, Canada
| | - Jean-François Yale
- The Research Institute of McGill University Health Centre, Montréal, Québec, Canada; Royal Victoria Hospital, McGill University Health Centre, Montréal, Québec, Canada
| | - Anas El Fathi
- Department of Biomedical Engineering, McGill University, Montréal, Québec, Canada
| | - Joanna Rutkowski
- Department of Biomedical Engineering, McGill University, Montréal, Québec, Canada
| | - Jennifer Rene
- Royal Victoria Hospital, McGill University Health Centre, Montréal, Québec, Canada
| | - Natasha Garfield
- Royal Victoria Hospital, McGill University Health Centre, Montréal, Québec, Canada
| | - Laurent Legault
- The Research Institute of McGill University Health Centre, Montréal, Québec, Canada; Montreal Children's Hospital, McGill University Health Centre, Montréal, Québec, Canada
| | - Ahmad Haidar
- Department of Biomedical Engineering, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Insulin is the mainstay of treatment in people living with type 1 diabetes mellitus due to an immune-mediated loss of beta cells. Yet despite advances in insulin therapy and other technological advances, glycemic control remains difficult to achieve. Therefore, we aim to highlight risks and benefits of adjunctive therapies that may improve type 1 diabetes care. RECENT FINDINGS We identified studies assessing clinical outcomes of adjunctive therapies that are both Food and Drug Administration (FDA)-approved and off-label in type 1 diabetes. Adjunctive therapies reviewed included metformin, pramlintide, glucagon-like peptide-1 (GLP-1) receptor agonists and sodium-glucose cotransporter-2 inhibitors. SUMMARY Although insulin is required in people living with type 1 diabetes mellitus, adjunctive therapies may positively impact glycemic control, reduce insulin requirements and lead to weight loss. In addition, the risk of hypoglycemia, gastrointestinal side effects and diabetes ketoacidosis may be increased with the use of these adjunctive therapies. Pramlintide is currently the only FDA-approved adjunctive therapy, whereas others require continued research to better understand risk-to-benefit ratio.
Collapse
Affiliation(s)
- Kyrstin Lane
- Division of Endocrinology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | | |
Collapse
|
20
|
Abstract
Insulin therapy has advanced remarkably over the past few decades. Ultra-rapid-acting and ultra-long-acting insulin analogs are now commercially available. Many additional insulin formulations are in development. This review outlines recent advances in insulin therapy and novel therapies in development.
Collapse
Affiliation(s)
- Leah M. Wilson
- Division of Endocrinology, Harold Schnitzer Diabetes Health Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Jessica R. Castle
- Division of Endocrinology, Harold Schnitzer Diabetes Health Center, Oregon Health & Science University, Portland, Oregon, USA
- Address correspondence to: Jessica R. Castle, MD, Division of Endocrinology, Harold Schnitzer Diabetes Health Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, L607, Portland, OR 97239-3098, USA
| |
Collapse
|
21
|
Glucose Control During Physical Activity and Exercise Using Closed Loop Technology in Adults and Adolescents with Type 1 Diabetes. Can J Diabetes 2020; 44:740-749. [DOI: 10.1016/j.jcjd.2020.06.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022]
|
22
|
Galderisi A, Cohen N, Calhoun P, Kraemer K, Breton M, Weinzimer S, Cengiz E. Effect of Afrezza on Glucose Dynamics During HCL Treatment. Diabetes Care 2020; 43:2146-2152. [PMID: 32661108 PMCID: PMC7440894 DOI: 10.2337/dc20-0091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/15/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE A major obstacle in optimizing the performance of closed-loop automated insulin delivery systems has been the delay in insulin absorption and action that results from the subcutaneous (SC) route of insulin delivery leading to exaggerated postmeal hyperglycemic excursions. We aimed to investigate the effect of Afrezza inhaled insulin with ultrafast-in and -out action profile on improving postprandial blood glucose control during hybrid closed-loop (HCL) treatment in young adults with type 1 diabetes. RESEARCH DESIGN AND METHODS We conducted an inpatient, three-way, randomized crossover standardized meal study to assess the efficacy and safety of Afrezza at a low (AL) and a high (AH) dose as compared with a standard SC rapid-acting insulin (aspart) premeal bolus during Diabetes Assistant (DiAs) HCL treatment. Participants received two sequential meals on three study days, and premeal insulin bolus was determined based on home insulin-to-carbohydrate ratio for each meal (rounded up to the closest available Afrezza cartridge dose for AH and down for AL). The primary efficacy outcome was the peak postprandial plasma glucose (PPG) level calculated by pooling data for up to 4 h after the start of each meal. Secondary outcomes included hyperglycemic, hypoglycemic, and euglycemic venous glucose metrics. RESULTS The mean ± SD PPG for the rapid-acting insulin control arm and AH was similar (185 ± 50 mg/dL vs. 195 ± 46 mg/dL, respectively; P = 0.45), while it was higher for meals using AL (208 ± 54 mg/dL, P = 0.04). The AH achieved significantly lower early PPG level than the control arm (30 min; P < 0.001), and improvement in PPG waned at later time points (120 and 180 min; P = 0.02) coinciding with the end of Afrezza glucodynamic action. CONCLUSIONS Afrezza (AH) premeal bolus reduced the early glycemic excursion and improved PPG during HCL compared with aspart premeal bolus. The improvement in PPG was not sustained after the end of Afrezza glucodynamic action at 120 min.
Collapse
Affiliation(s)
- Alfonso Galderisi
- Department of Pediatrics, Yale School of Medicine, New Haven, CT .,Department of Women's and Children's Health, University of Padova, Padova, Italy
| | | | | | - Kristen Kraemer
- Department of Pediatrics, Yale School of Medicine, New Haven, CT
| | - Marc Breton
- Center for Diabetes Technology, University of Virginia, Charlottesville, VA
| | - Stuart Weinzimer
- Department of Pediatrics, Yale School of Medicine, New Haven, CT
| | - Eda Cengiz
- Department of Pediatrics, Yale School of Medicine, New Haven, CT.,Bahcesehir University School of Medicine, Istanbul, Turkey
| |
Collapse
|
23
|
Abstract
Treatments for type 1 diabetes have advanced significantly over recent years. There are now multiple hybrid closed-loop systems commercially available and additional systems are in development. Challenges remain, however. This review outlines the recent advances in closed-loop systems and outlines the remaining challenges, including post-prandial hyperglycemia and exercise-related dysglycemia.
Collapse
Affiliation(s)
- Melanie Jackson
- Division of Endocrinology, Harold Schnitzer Diabetes Health Center, Oregon Health & Science University, Portland, Oregon
| | - Jessica R. Castle
- Division of Endocrinology, Harold Schnitzer Diabetes Health Center, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
24
|
Maikawa CL, Smith AAA, Zou L, Roth GA, Gale EC, Stapleton LM, Baker SW, Mann JL, Yu AC, Correa S, Grosskopf AK, Liong CS, Meis CM, Chan D, Troxell M, Maahs DM, Buckingham BA, Webber MJ, Appel EA. A co-formulation of supramolecularly stabilized insulin and pramlintide enhances mealtime glucagon suppression in diabetic pigs. Nat Biomed Eng 2020; 4:507-517. [PMID: 32393892 PMCID: PMC7274092 DOI: 10.1038/s41551-020-0555-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/03/2020] [Indexed: 02/06/2023]
Abstract
Treatment of patients with diabetes with insulin and pramlintide (an amylin analogue) is more effective than treatment with insulin only. However, because mixtures of insulin and pramlintide are unstable and have to be injected separately, amylin analogues are only used by 1.5% of people with diabetes needing rapid-acting insulin. Here, we show that the supramolecular modification of insulin and pramlintide with cucurbit[7]uril-conjugated polyethylene glycol improves the pharmacokinetics of the dual-hormone therapy and enhances postprandial glucagon suppression in diabetic pigs. The co-formulation is stable for over 100 h at 37 °C under continuous agitation, whereas commercial formulations of insulin analogues aggregate after 10 h under similar conditions. In diabetic rats, the administration of the stabilized co-formulation increased the area-of-overlap ratio of the pharmacokinetic curves of pramlintide and insulin from 0.4 ± 0.2 to 0.7 ± 0.1 (mean ± s.d.) for the separate administration of the hormones. The co-administration of supramolecularly stabilized insulin and pramlintide better mimics the endogenous kinetics of co-secreted insulin and amylin, and holds promise as a dual-hormone replacement therapy.
Collapse
Affiliation(s)
- Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Anton A A Smith
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Department of Science and Technology, Aarhus University, Aarhus, Denmark
| | - Lei Zou
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Gillie A Roth
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Emily C Gale
- Department of Biochemistry, Stanford University, Stanford, CA, USA
| | | | - Sam W Baker
- Department of Comparative Medicine, Stanford University, Stanford, CA, USA
| | - Joseph L Mann
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Anthony C Yu
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Santiago Correa
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | | | - Celine S Liong
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Catherine M Meis
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Doreen Chan
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Megan Troxell
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - David M Maahs
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, USA
- Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Bruce A Buckingham
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, USA
- Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Matthew J Webber
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
- Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA, USA.
- Diabetes Research Center, Stanford University, Stanford, CA, USA.
| |
Collapse
|
25
|
Haidar A, Tsoukas MA, Bernier-Twardy S, Yale JF, Rutkowski J, Bossy A, Pytka E, El Fathi A, Strauss N, Legault L. A Novel Dual-Hormone Insulin-and-Pramlintide Artificial Pancreas for Type 1 Diabetes: A Randomized Controlled Crossover Trial. Diabetes Care 2020; 43:597-606. [PMID: 31974099 DOI: 10.2337/dc19-1922] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/22/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The rapid insulin-alone artificial pancreas improves glycemia in type 1 diabetes but daytime control remains suboptimal. We propose two novel dual-hormone artificial pancreas systems. RESEARCH DESIGN AND METHODS We conducted a randomized crossover trial comparing a rapid insulin-alone artificial pancreas with rapid insulin-and-pramlintide and with regular insulin-and-pramlintide artificial pancreas systems in adults with type 1 diabetes. Participants were assigned to the interventions in random order during three 24-h inpatient visits. Each visit was preceded by an outpatient hormonal open-loop run-in period of 10-14 days. The dual-hormone artificial pancreas delivered pramlintide in a basal-bolus manner, using a novel dosing algorithm, with a fixed ratio relative to insulin. The primary outcome was time in the range 3.9-10.0 mmol/L. RESULTS Compared with the rapid insulin-alone artificial pancreas system, the rapid insulin-and-pramlintide system increased the time in range from 74% (SD 18%) to 84% (13%) (P = 0.0014), whereas the regular insulin-and-pramlintide system did not change the time in range (69% [19%]; P = 0.22). The increased time in range with the rapid insulin-and-pramlintide system was due to improved daytime control (daytime time in range increased from 63% [23%] to 78% [16%], P = 0.0004). There were 11 (1 per 2.5 days) hypoglycemic events (<3.3 mmol/L with symptoms or <3.0 mmol/L irrespective of symptoms) with the rapid insulin-alone system, compared with 12 (1 per 2.3 days) and 18 (1 per 1.4 days) with the rapid and regular insulin-and-pramlintide systems, respectively. Gastrointestinal symptoms were reported after 0% (0 of 112) of meals with the rapid insulin-alone system, compared with 6% (6 of 108) and 11% (11 of 104) with the rapid and regular insulin-and-pramlintide systems, respectively; none of the symptoms were severe. CONCLUSIONS A novel rapid insulin-and-pramlintide artificial pancreas improves glucose control compared with a rapid insulin-alone artificial pancreas (ClinicalTrials.gov number NCT02814123).
Collapse
Affiliation(s)
- Ahmad Haidar
- Department of Biomedical Engineering, McGill University, Montréal, Québec, Canada .,The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - Michael A Tsoukas
- The Research Institute of McGill University Health Centre, Montréal, Québec, Canada.,Royal Victoria Hospital, McGill University Health Centre, Montréal, Québec, Canada
| | - Sarah Bernier-Twardy
- Department of Biomedical Engineering, McGill University, Montréal, Québec, Canada
| | - Jean-Francois Yale
- The Research Institute of McGill University Health Centre, Montréal, Québec, Canada.,Royal Victoria Hospital, McGill University Health Centre, Montréal, Québec, Canada
| | - Joanna Rutkowski
- Department of Biomedical Engineering, McGill University, Montréal, Québec, Canada
| | - Anne Bossy
- Department of Biomedical Engineering, McGill University, Montréal, Québec, Canada
| | - Evelyne Pytka
- Department of Biomedical Engineering, McGill University, Montréal, Québec, Canada
| | - Anas El Fathi
- Department of Biomedical Engineering, McGill University, Montréal, Québec, Canada
| | - Natalia Strauss
- Department of Biomedical Engineering, McGill University, Montréal, Québec, Canada
| | - Laurent Legault
- Montreal Children's Hospital, McGill University Health Centre, Montréal, Québec, Canada
| |
Collapse
|
26
|
Abstract
Treatment of type 1 diabetes with exogenous insulin often results in unpredictable daily glucose variability and hypoglycemia, which can be dangerous. Automated insulin delivery systems can improve glucose control while reducing burden for people with diabetes. One approach to improve treatment outcomes is to incorporate the counter-regulatory hormone glucagon into the automated delivery system to help prevent the hypoglycemia that can be induced by the slow pharmacodynamics of insulin action. This article explores the advantages and disadvantages of incorporating glucagon into dual-hormone automated hormone delivery systems.
Collapse
Affiliation(s)
- Leah M Wilson
- Division of Endocrinology, Diabetes and Clinical Nutrition, Oregon Health & Science University, Harold Schnitzer Diabetes Health Center, 3181 Southwest Sam Jackson Park Road, L607, Portland, OR 97239-3098, USA.
| | - Peter G Jacobs
- Department of Biomedical Engineering, Oregon Health & Science University, Mail Code: CH13B, 3303 Southwest Bond Avenue, Portland, OR 97239, USA
| | - Jessica R Castle
- Division of Endocrinology, Diabetes and Clinical Nutrition, Oregon Health & Science University, Harold Schnitzer Diabetes Health Center, 3181 Southwest Sam Jackson Park Road, L607, Portland, OR 97239-3098, USA
| |
Collapse
|
27
|
Lal RA, Ekhlaspour L, Hood K, Buckingham B. Realizing a Closed-Loop (Artificial Pancreas) System for the Treatment of Type 1 Diabetes. Endocr Rev 2019; 40:1521-1546. [PMID: 31276160 PMCID: PMC6821212 DOI: 10.1210/er.2018-00174] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 02/28/2019] [Indexed: 01/20/2023]
Abstract
Recent, rapid changes in the treatment of type 1 diabetes have allowed for commercialization of an "artificial pancreas" that is better described as a closed-loop controller of insulin delivery. This review presents the current state of closed-loop control systems and expected future developments with a discussion of the human factor issues in allowing automation of glucose control. The goal of these systems is to minimize or prevent both short-term and long-term complications from diabetes and to decrease the daily burden of managing diabetes. The closed-loop systems are generally very effective and safe at night, have allowed for improved sleep, and have decreased the burden of diabetes management overnight. However, there are still significant barriers to achieving excellent daytime glucose control while simultaneously decreasing the burden of daytime diabetes management. These systems use a subcutaneous continuous glucose sensor, an algorithm that accounts for the current glucose and rate of change of the glucose, and the amount of insulin that has already been delivered to safely deliver insulin to control hyperglycemia, while minimizing the risk of hypoglycemia. The future challenge will be to allow for full closed-loop control with minimal burden on the patient during the day, alleviating meal announcements, carbohydrate counting, alerts, and maintenance. The human factors involved with interfacing with a closed-loop system and allowing the system to take control of diabetes management are significant. It is important to find a balance between enthusiasm and realistic expectations and experiences with the closed-loop system.
Collapse
Affiliation(s)
- Rayhan A Lal
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, California.,Division of Endocrinology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Laya Ekhlaspour
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Korey Hood
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, California.,Department of Psychiatry, Stanford University School of Medicine, Stanford, California
| | - Bruce Buckingham
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
28
|
van Huijgevoort NCM, Del Chiaro M, Wolfgang CL, van Hooft JE, Besselink MG. Diagnosis and management of pancreatic cystic neoplasms: current evidence and guidelines. Nat Rev Gastroenterol Hepatol 2019; 16:676-689. [PMID: 31527862 DOI: 10.1038/s41575-019-0195-x] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/31/2019] [Indexed: 12/11/2022]
Abstract
Pancreatic cystic neoplasms (PCN) are a heterogeneous group of pancreatic cysts that include intraductal papillary mucinous neoplasms, mucinous cystic neoplasms, serous cystic neoplasms and other rare cystic lesions, all with different biological behaviours and variable risk of progression to malignancy. As more pancreatic cysts are incidentally discovered on routine cross-sectional imaging, optimal surveillance for patients with PCN is becoming an increasingly common clinical problem, highlighting the need to balance cancer prevention with the risk of (surgical) overtreatment. This Review summarizes the latest developments in the diagnosis and management of PCN, including the quality of available evidence. Also discussed are the most important differences between the PCN guidelines from the American Gastroenterological Association, the International Association of Pancreatology and the European Study Group on Cystic Tumours of the Pancreas, including diagnostic and follow-up strategies and indications for surgery. Finally, new developments in the management of patients with PCN are addressed.
Collapse
Affiliation(s)
- Nadine C M van Huijgevoort
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Marco Del Chiaro
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Christopher L Wolfgang
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Marc G Besselink
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
29
|
Haidar A. Insulin-and-Glucagon Artificial Pancreas Versus Insulin-Alone Artificial Pancreas: A Short Review. Diabetes Spectr 2019; 32:215-221. [PMID: 31462876 PMCID: PMC6695257 DOI: 10.2337/ds18-0097] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
IN BRIEF The advantage of the insulin-and-glucagon artificial pancreas is based on the rapid effect of subcutaneous glucagon delivery in preventing hypoglycemia compared to suspension of insulin delivery. In short-term studies, the dual-hormone artificial pancreas reduced daytime hypoglycemia, especially during exercise, compared to the insulin-alone artificial pancreas, but the insulin-alone system seemed sufficient in eliminating nocturnal hypoglycemia. The comparative benefits of the single- and dual-hormone systems for improving A1C and preventing severe hypoglycemia remain unknown.
Collapse
Affiliation(s)
- Ahmad Haidar
- Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
- Division of Endocrinology and Metabolism, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
30
|
Artificial Pancreas: Current Progress and Future Outlook in the Treatment of Type 1 Diabetes. Drugs 2019; 79:1089-1101. [DOI: 10.1007/s40265-019-01149-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
31
|
Nally LM, Sherr JL, Van Name MA, Patel AD, Tamborlane WV. Pharmacologic treatment options for type 1 diabetes: what's new? Expert Rev Clin Pharmacol 2019; 12:471-479. [PMID: 30892094 DOI: 10.1080/17512433.2019.1597705] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The expanding variety of insulins, including biosynthetic human insulin and rapid and long-acting insulin analogs, have dramatically transformed the management of type 1 diabetes (T1D) over the past 25 years. Moreover, increasing interest in the use of novel drugs developed for the treatment of type 2 diabetes (T2D) as adjunctive therapies for T1D remains a work in progress. Areas Covered: We reviewed articles published up to December 2018 in PubMed and ClinicalTrials.gov for recent developments in the pharmacologic treatment of T1D, including inhaled insulin, ultrafast and ultralong-acting insulins and adjunctive therapies including pramlintide, metformin, GLP-1 receptor agonists, DPP-4 inhibitors, SGLT-2, and SGLT1/2 inhibitors. Expert Opinion: With the creation of ultrafast-acting insulin analogs and very prolonged duration of action of basal insulins, it is possible to more closely mimic physiologic insulin secretion. Adjunctive therapies, likewise, may also overcome some of the abnormal physiology that is a hallmark of T1D. Therefore, individualized consideration of the efficacy of these agents must be measured alongside the potential adverse effects when choosing an adjunctive therapy.
Collapse
Affiliation(s)
- Laura M Nally
- a Yale Children's Diabetes Program , Yale University School of Medicine , New Haven , CT , USA
| | - Jennifer L Sherr
- a Yale Children's Diabetes Program , Yale University School of Medicine , New Haven , CT , USA
| | - Michelle A Van Name
- a Yale Children's Diabetes Program , Yale University School of Medicine , New Haven , CT , USA
| | - Anisha D Patel
- a Yale Children's Diabetes Program , Yale University School of Medicine , New Haven , CT , USA
| | - William V Tamborlane
- a Yale Children's Diabetes Program , Yale University School of Medicine , New Haven , CT , USA
| |
Collapse
|
32
|
Navigating Two Roads to Glucose Normalization in Diabetes: Automated Insulin Delivery Devices and Cell Therapy. Cell Metab 2019; 29:545-563. [PMID: 30840911 DOI: 10.1016/j.cmet.2019.02.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 12/23/2022]
Abstract
Incredible strides have been made since the discovery of insulin almost 100 years ago. Insulin formulations have improved dramatically, glucose levels can be measured continuously, and recently first-generation biomechanical "artificial pancreas" systems have been approved by regulators around the globe. However, still only a small fraction of patients with diabetes achieve glycemic goals. Replacement of insulin-producing cells via transplantation shows significant promise, but is limited in application due to supply constraints (cadaver-based) and the need for chronic immunosuppression. Over the past decade, significant progress has been made to address these barriers to widespread implementation of a cell therapy. Can glucose levels in people with diabetes be normalized with artificial pancreas systems or via cell replacement approaches? Here we review the road ahead, including the challenges and opportunities of both approaches.
Collapse
|
33
|
Zhang XX, Qiao YC, Li W, Zou X, Chen YL, Shen J, Liao QY, Zhang QJ, He L, Zhao HL. Human amylin induces CD4+Foxp3+ regulatory T cells in the protection from autoimmune diabetes. Immunol Res 2019; 66:179-186. [PMID: 28983871 DOI: 10.1007/s12026-017-8956-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Autoimmune diabetes is a disorder of immune homeostasis that leads to targeted insulin-secreting islet β cell destruction characterized by insulitis. Human amylin (hA) is an important neuroendocrine hormone co-secreted with insulin by pancreatic β cells. Here, we report hA immune-modulatory action through inducing regulatory T cells. We ex vivo-treated human peripheral blood mononuclear cells (hPBMCs) with hA for 24 h and counted CD4+Foxp3+ regulatory T cells (Treg) using flow cytometry. Diabetic status was monitored and splenic Treg were measured in non-obese diabetic (NOD) male mice. NOD mice were intraperitoneally injected once daily with hA (n = 25) or solvent for control (n = 25) for 7 months continuously. Spleen tissues were collected at the end of intervention and processed for flow cytometry and Western blot. We found a 2.9-fold (p < 0.05) increase of CD4+Foxp3+ Treg in hPBMCs treated with 10 nmol/L hA compared with negative control. Incidence of diabetes in hA-treated NOD mice decreased 44% (p = 0.045) in the 6th month and 57% (p = 0.0002) in the 7th month. Meanwhile, the hA treatment induced a 1.5-fold increase of CD4+Foxp3+ Treg from mouse splenocytes (p = 0.0013). Expression of transforming growth factor-β (TGF-β) and toll-like receptor-4 (TLR-4) were upregulated in hA-treated mice. Human amylin might protect against autoimmune diabetes via the induction of CD4+Foxp3+ Treg, which suggests a novel approach to improve autoimmune conditions.
Collapse
Affiliation(s)
- Xiao-Xi Zhang
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Yong-Chao Qiao
- Department of Immunology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Wan Li
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Xia Zou
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Yin-Ling Chen
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Jian Shen
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Qin-Yuan Liao
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Qiu-Jin Zhang
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Lan He
- Department of Microbiology, Prince of Wales Hospital, Shatin, Hong Kong
| | - Hai-Lu Zhao
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China. .,Department of Immunology, Guilin Medical University, Guilin, Guangxi, 541004, China.
| |
Collapse
|
34
|
Karageorgiou V, Papaioannou TG, Bellos I, Alexandraki K, Tentolouris N, Stefanadis C, Chrousos GP, Tousoulis D. Effectiveness of artificial pancreas in the non-adult population: A systematic review and network meta-analysis. Metabolism 2019; 90:20-30. [PMID: 30321535 DOI: 10.1016/j.metabol.2018.10.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/20/2018] [Accepted: 10/09/2018] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Artificial pancreas is a technology that minimizes user input by bridging continuous glucose monitoring and insulin pump treatment, and has proven safety in the adult population. The purpose of this systematic review and meta-analysis is to evaluate the efficacy of closed-loop (CL) systems in the glycemic control of non-adult type 1 diabetes patients in both a pairwise and network meta-analysis (NMA) context and investigate various parameters potentially affecting the outcome. METHODS Literature was systematically searched using the MEDLINE (1966-2018), Scopus (2004-2018), Cochrane Central Register of Controlled Trials (CENTRAL) (1999-2018), Clinicaltrials.gov (2008-2018) and Google Scholar (2004-2018) databases. Studies comparing the glycemic control in CL (either single- or dual-hormone) with continuous subcutaneous insulin infusion (CSII) in people with diabetes (PWD) aged <18 years old were deemed eligible. The primary outcome analysis was conducted with regard to time spent in the target glycemic range. All outcomes were evaluated in NMA in order to investigate potential between-algorithm differences. Pairwise meta-analysis and meta-regression were performed using the RevMan 5.3 and Open Meta-Analyst software. For NMA, the package pcnetmetain R 3.5.1 was used. RESULTS The meta-analysis was based on 25 studies with a total of 504 PWD. The CL group was associated with significantly higher percentage of time spent in the target glycemic range (Mean (SD): 67.59% (SD: 8.07%) in the target range and OL PWD spending 55.77% (SD: 11.73%), MD: -11.97%, 95% CI [-18.40, -5.54%]) and with lower percentages of time in hyperglycemia (MD: 3.01%, 95% CI [1.68, 4.34%]) and hypoglycemia (MD: 0.67%, 95% CI [0.21, 1.13%]. Mean glucose was also decreased in the CL group (MD: 0.75 mmol/L, 95% CI [0.18-1.33]). The NMA arm of the study showed that the bihormonal modality was superior to other algorithms and standard treatment in lowering mean glucose and increasing time spent in the target range. The DiAs platform was superior to PID in controlling hypoglycemia and mean glucose. Time in target range and mean glucose were unaffected by the confounding factors tested. CONCLUSIONS The findings of this meta-analysis suggest that artificial pancreas systems are superior to the standard sensor-augmented pump treatment of type 1 diabetes mellitus in non-adult PWD. Between-algorithm differences are also addressed, implying a superiority of the bihormonal treatment modality. Future large-scale studies are needed in the field to verify these outcomes and to determine the optimal algorithm to be used in the clinical setting.
Collapse
Affiliation(s)
- Vasilios Karageorgiou
- First Department of Cardiology, Biomedical Engineering Unit, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros G Papaioannou
- First Department of Cardiology, Biomedical Engineering Unit, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - Ioannis Bellos
- First Department of Cardiology, Biomedical Engineering Unit, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Krystallenia Alexandraki
- Clinic of Endocrine Oncology, Section of Endocrinology, Department of Pathophysiology, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Tentolouris
- First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - George P Chrousos
- First Department of Pediatrics, Aghia Sophia Children's Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Tousoulis
- First Department of Cardiology, Biomedical Engineering Unit, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
35
|
Müller TD, Clemmensen C, Finan B, DiMarchi RD, Tschöp MH. Anti-Obesity Therapy: from Rainbow Pills to Polyagonists. Pharmacol Rev 2018; 70:712-746. [PMID: 30087160 DOI: 10.1124/pr.117.014803] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
With their ever-growing prevalence, obesity and diabetes represent major health threats of our society. Based on estimations by the World Health Organization, approximately 300 million people will be obese in 2035. In 2015 alone there were more than 1.6 million fatalities attributable to hyperglycemia and diabetes. In addition, treatment of these diseases places an enormous burden on our health care system. As a result, the development of pharmacotherapies to tackle this life-threatening pandemic is of utmost importance. Since the beginning of the 19th century, a variety of drugs have been evaluated for their ability to decrease body weight and/or to improve deranged glycemic control. The list of evaluated drugs includes, among many others, sheep-derived thyroid extracts, mitochondrial uncouplers, amphetamines, serotonergics, lipase inhibitors, and a variety of hormones produced and secreted by the gastrointestinal tract or adipose tissue. Unfortunately, when used as a single hormone therapy, most of these drugs are underwhelming in their efficacy or safety, and placebo-subtracted weight loss attributed to such therapy is typically not more than 10%. In 2009, the generation of a single molecule with agonism at the receptors for glucagon and the glucagon-like peptide 1 broke new ground in obesity pharmacology. This molecule combined the beneficial anorectic and glycemic effects of glucagon-like peptide 1 with the thermogenic effect of glucagon into a single molecule with enhanced potency and sustained action. Several other unimolecular dual agonists have subsequently been developed, and, based on their preclinical success, these molecules illuminate the path to a new and more fruitful era in obesity pharmacology. In this review, we focus on the historical pharmacological approaches to treat obesity and glucose intolerance and describe how the knowledge obtained by these studies led to the discovery of unimolecular polypharmacology.
Collapse
Affiliation(s)
- T D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany (T.D.M., C.C., M.H.T.); German Center for Diabetes Research, Neuherberg, Germany (T.D.M., C.C., M.H.T.); Department of Chemistry, Indiana University, Bloomington, Indiana (B.F., R.D.D.); and Division of Metabolic Diseases, Technische Universität München, Munich, Germany (M.H.T.)
| | - C Clemmensen
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany (T.D.M., C.C., M.H.T.); German Center for Diabetes Research, Neuherberg, Germany (T.D.M., C.C., M.H.T.); Department of Chemistry, Indiana University, Bloomington, Indiana (B.F., R.D.D.); and Division of Metabolic Diseases, Technische Universität München, Munich, Germany (M.H.T.)
| | - B Finan
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany (T.D.M., C.C., M.H.T.); German Center for Diabetes Research, Neuherberg, Germany (T.D.M., C.C., M.H.T.); Department of Chemistry, Indiana University, Bloomington, Indiana (B.F., R.D.D.); and Division of Metabolic Diseases, Technische Universität München, Munich, Germany (M.H.T.)
| | - R D DiMarchi
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany (T.D.M., C.C., M.H.T.); German Center for Diabetes Research, Neuherberg, Germany (T.D.M., C.C., M.H.T.); Department of Chemistry, Indiana University, Bloomington, Indiana (B.F., R.D.D.); and Division of Metabolic Diseases, Technische Universität München, Munich, Germany (M.H.T.)
| | - M H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany (T.D.M., C.C., M.H.T.); German Center for Diabetes Research, Neuherberg, Germany (T.D.M., C.C., M.H.T.); Department of Chemistry, Indiana University, Bloomington, Indiana (B.F., R.D.D.); and Division of Metabolic Diseases, Technische Universität München, Munich, Germany (M.H.T.)
| |
Collapse
|
36
|
Sherr JL, Tauschmann M, Battelino T, de Bock M, Forlenza G, Roman R, Hood KK, Maahs DM. ISPAD Clinical Practice Consensus Guidelines 2018: Diabetes technologies. Pediatr Diabetes 2018; 19 Suppl 27:302-325. [PMID: 30039513 DOI: 10.1111/pedi.12731] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Jennifer L Sherr
- Department of Pediatrics, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Martin Tauschmann
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.,Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Tadej Battelino
- UMC-University Children's Hospital, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Martin de Bock
- Department of Paediatrics, University of Otago, Christchurch, New Zealand
| | - Gregory Forlenza
- University of Colorado Denver, Barbara Davis Center, Aurora, Colorado
| | - Rossana Roman
- Medical Sciences Department, University of Antofagasta and Antofagasta Regional Hospital, Antofagasta, Chile
| | - Korey K Hood
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California
| | - David M Maahs
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California
| |
Collapse
|
37
|
Corbin KD, Driscoll KA, Pratley RE, Smith SR, Maahs DM, Mayer-Davis EJ. Obesity in Type 1 Diabetes: Pathophysiology, Clinical Impact, and Mechanisms. Endocr Rev 2018; 39:629-663. [PMID: 30060120 DOI: 10.1210/er.2017-00191] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 06/21/2018] [Indexed: 02/07/2023]
Abstract
There has been an alarming increase in the prevalence of obesity in people with type 1 diabetes in recent years. Although obesity has long been recognized as a major risk factor for the development of type 2 diabetes and a catalyst for complications, much less is known about the role of obesity in the initiation and pathogenesis of type 1 diabetes. Emerging evidence suggests that obesity contributes to insulin resistance, dyslipidemia, and cardiometabolic complications in type 1 diabetes. Unique therapeutic strategies may be required to address these comorbidities within the context of intensive insulin therapy, which promotes weight gain. There is an urgent need for clinical guidelines for the prevention and management of obesity in type 1 diabetes. The development of these recommendations will require a transdisciplinary research strategy addressing metabolism, molecular mechanisms, lifestyle, neuropsychology, and novel therapeutics. In this review, the prevalence, clinical impact, energy balance physiology, and potential mechanisms of obesity in type 1 diabetes are described, with a special focus on the substantial gaps in knowledge in this field. Our goal is to provide a framework for the evidence base needed to develop type 1 diabetes-specific weight management recommendations that account for the competing outcomes of glycemic control and weight management.
Collapse
Affiliation(s)
- Karen D Corbin
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - Kimberly A Driscoll
- Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, Colorado.,Barbara Davis Center for Diabetes, Aurora, Colorado
| | - Richard E Pratley
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - David M Maahs
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California
| | - Elizabeth J Mayer-Davis
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | |
Collapse
|
38
|
Tauschmann M, Hovorka R. Technology in the management of type 1 diabetes mellitus - current status and future prospects. Nat Rev Endocrinol 2018; 14:464-475. [PMID: 29946127 DOI: 10.1038/s41574-018-0044-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Type 1 diabetes mellitus (T1DM) represents 5-10% of diabetes cases worldwide. The incidence of T1DM is increasing, and there is no immediate prospect of a cure. As such, lifelong management is required, the burden of which is being eased by novel treatment modalities, particularly from the field of diabetes technologies. Continuous glucose monitoring has become the standard of care and includes factory-calibrated subcutaneous glucose monitoring and long-term implantable glucose sensing. In addition, considerable progress has been made in technology-enabled glucose-responsive insulin delivery. The first hybrid insulin-only closed-loop system has been commercialized, and other closed-loop systems are under development, including dual-hormone glucose control systems. This Review focuses on well-established diabetes technologies, including glucose sensing, pen-based insulin delivery, data management and data analytics. We also cover insulin pump therapy, threshold-based suspend, predictive low-glucose suspend and single-hormone and dual-hormone closed-loop systems. Clinical practice recommendations for insulin pump therapy and continuous glucose monitoring are presented, and ongoing research and future prospects are highlighted. We conclude that the management of T1DM is improved by diabetes technology for the benefit of the majority of people with T1DM, their caregivers and guardians and health-care professionals treating patients with T1DM.
Collapse
Affiliation(s)
- Martin Tauschmann
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Roman Hovorka
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Department of Paediatrics, University of Cambridge, Cambridge, UK.
| |
Collapse
|
39
|
Sherr JL. Closing the Loop on Managing Youth With Type 1 Diabetes: Children Are Not Just Small Adults. Diabetes Care 2018; 41:1572-1578. [PMID: 29936422 PMCID: PMC6054496 DOI: 10.2337/dci18-0003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 04/24/2018] [Indexed: 02/03/2023]
Abstract
As hybrid closed-loop (HCL) insulin delivery systems permeate clinical practice, it is critical to ensure all with diabetes are afforded the opportunity to benefit from this technology. Indeed, due to the suboptimal control achieved by the vast majority of youth with type 1 diabetes (T1D), pediatric patients are positioned to see the greatest benefit from automated insulin delivery systems. To ensure these systems are well poised to deliver the promise of more targeted control, it is essential to understand the unique characteristics and factors of childhood. Herein, the developmental and physiological needs of youth with T1D are reviewed and consideration is given to how HCL could address these issues. Studies of HCL technologies in youth are briefly reviewed. As future-generation closed-loop systems are being devised, features that could make this technology more attractive to youth and to their families are discussed. Integration of HCL has the potential to minimize the burden of this chronic medical condition while improving glycemic control and ultimately allowing our pediatric patients to fulfill the primary goal of childhood, to be a kid.
Collapse
Affiliation(s)
- Jennifer L Sherr
- Pediatric Endocrinology & Diabetes Section, Department of Pediatrics, Yale School of Medicine, New Haven, CT
| |
Collapse
|
40
|
Majeed W, Thabit H. Closed-loop insulin delivery: current status of diabetes technologies and future prospects. Expert Rev Med Devices 2018; 15:579-590. [PMID: 30027775 DOI: 10.1080/17434440.2018.1503530] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Type 1 diabetes is characterised by destruction of pancreatic beta cells, leading to insulin deficiency and hyperglycaemia. The mainstay of treatment remains lifelong insulin therapy as a sustainable cure has as yet proven elusive. The burden of daily management of type 1 diabetes has contributed to suboptimal outcomes for people living with the condition. Innovative technological approaches have been shown to improve glycaemic and patient-related outcomes. AREAS COVERED We discuss recent advances in technologies in type 1 diabetes including closed-loop systems, also known as the 'artificial pancreas. Its various components, technical aspects and limitations are reviewed. We also discuss its advent into clinical practice, and other systems in development. Evidence from clinical studies are summarised. EXPERT COMMENTARY The recent approval of a hybrid closed-loop system for clinical use highlights the significant progress made in this field. Results from clinical studies have shown safety and glycaemic benefit, however challenges remain around improving performance and acceptability. More data is required to establish long-term clinical efficacy and cost-effectiveness, to fulfil the expectations of people with type 1 diabetes.
Collapse
Affiliation(s)
- Waseem Majeed
- a Manchester Academic Health Science Centre , Manchester University Hospitals NHS Foundation Trust , Manchester , UK
| | - Hood Thabit
- a Manchester Academic Health Science Centre , Manchester University Hospitals NHS Foundation Trust , Manchester , UK.,b Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health , University of Manchester , Manchester , UK
| |
Collapse
|
41
|
Esposito S, Santi E, Mancini G, Rogari F, Tascini G, Toni G, Argentiero A, Berioli MG. Efficacy and safety of the artificial pancreas in the paediatric population with type 1 diabetes. J Transl Med 2018; 16:176. [PMID: 29954380 PMCID: PMC6022450 DOI: 10.1186/s12967-018-1558-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 06/23/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Type 1 diabetes (DM1) is one of the most common chronic diseases in childhood and requires life-long insulin therapy and continuous health care support. An artificial pancreas (AP) or closed-loop system (CLS) have been developed with the aim of improving metabolic control without increasing the risk of hypoglycaemia in patients with DM1. As the impact of APs have been studied mainly in adults, the aim of this review is to evaluate the efficacy and safety of the AP in the paediatric population with DM1. MAIN BODY The real advantage of a CLS compared to last-generation sensor-augmented pumps is the gradual modulation of basal insulin infusion in response to glycaemic variations (towards both hyperglycaemia and hypoglycaemia), which has the aim of improving the proportion of time spent in the target glucose range and reducing the mean glucose level without increasing the risk of hypoglycaemia. Some recent studies demonstrated that also in children and adolescents an AP is able to reduce the frequency of hypoglycaemic events, an important limiting factor in reaching good metabolic control, particularly overnight. However, the advantages of the AP in reducing hyperglycaemia, increasing the time spent in the target glycaemic range and thus reducing glycated haemoglobin are less clear and require more clinical trials in the paediatric population, in particular in younger children. CONCLUSIONS Although the first results from bi-hormonal CLS are promising, long-term, head-to-head studies will have to prove their superiority over insulin-only approaches. More technological progress, the availability of more fast-acting insulin, further developments of algorithms that could improve glycaemic control after meals and physical activity are the most important challenges in reaching an optimal metabolic control with the use of the AP in children and adolescents.
Collapse
Affiliation(s)
- Susanna Esposito
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy.
| | - Elisa Santi
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Giulia Mancini
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Francesco Rogari
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Giorgia Tascini
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Giada Toni
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Alberto Argentiero
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Maria Giulia Berioli
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| |
Collapse
|
42
|
Geyer MC, Rayner CK, Horowitz M, Couper JJ. Targeting postprandial glycaemia in children with diabetes: Opportunities and challenges. Diabetes Obes Metab 2018; 20:766-774. [PMID: 29072820 DOI: 10.1111/dom.13141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/02/2017] [Accepted: 10/21/2017] [Indexed: 02/05/2023]
Abstract
Postprandial glycaemia makes a substantial contribution to overall glycaemic control in diabetes, particularly in patients whose preprandial glycaemia is relatively well controlled and glycated haemoglobin (HbA1c) only modestly elevated. Our review addresses the determinants of postprandial glycaemia and how it may be targeted therapeutically in children with diabetes. Postprandial glycaemia is influenced by preprandial glycaemia, macronutrients and their absorption, insulin delivery and sensitivity, the action of the enteroendocrine system, and the rate of gastric emptying. Contemporary continuous glucose monitoring systems reveal patterns of post prandial glycaemia and allow management to be guided more precisely. Delays in blood glucose determination, insulin delivery and its absorption remain challenges in the rapidly evolving closed loop continuous subcutaneous insulin and glucagon delivery systems developed for children with type 1 diabetes. Augmentation of the incretin system through nutritional preloads or incretin mimetics targets postprandial glycaemia by slowing gastric emptying as well as insulinotropic and glucagonostatic effects. These treatments are of particular relevance to children with type 2 diabetes. Following the development of targeted therapies in adults, postprandial blood glucose control will now be increasingly targeted in the treatment of diabetes in children.
Collapse
Affiliation(s)
- Myfanwy C Geyer
- Discipline of Paediatrics, University of Adelaide, Adelaide, Australia
- Department of Endocrinology and Diabetes, Women's and Children's Hospital, Adelaide, Australia
| | - Christopher K Rayner
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, Australia
| | - Michael Horowitz
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, Australia
| | - Jennifer J Couper
- Department of Endocrinology and Diabetes, Women's and Children's Hospital, Adelaide, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
| |
Collapse
|
43
|
Peters TM, Haidar A. Dual-hormone artificial pancreas: benefits and limitations compared with single-hormone systems. Diabet Med 2018; 35:450-459. [PMID: 29337384 DOI: 10.1111/dme.13581] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2018] [Indexed: 12/17/2022]
Abstract
Technological advances have made the artificial pancreas a reality. This has the potential to improve the lives of individuals with Type 1 diabetes by reducing the risk of hypoglycaemia, achieving better overall glucose control, and enhancing quality of life. Both single-hormone (insulin-only) and dual-hormone (insulin and glucagon) systems have been developed; however, a focused review of the relative benefits of each artificial pancreas system is needed. We reviewed studies that directly compared single- and dual-hormone systems to evaluate the efficacy of each system for preventing hypoglycaemia and maintaining glycaemic control, as well as their utility in specific situations including during exercise, overnight and during the prandial period. We observed additional benefits with the dual-hormone artificial pancreas for reducing the risk of hypoglycaemic events overall and during exercise over the study duration. The single-hormone artificial pancreas was sufficient for maintenance of euglycaemia in the overnight period and for preventing late-onset post-exercise hypoglycaemia. Future comparative studies of longer duration are required to determine whether one system is superior for improving mean glucose control, eliminating severe hypoglycaemia, or improving quality of life.
Collapse
Affiliation(s)
- T M Peters
- Division of Endocrinology and Metabolism, Faculty of Medicine
| | - A Haidar
- Division of Endocrinology and Metabolism, Faculty of Medicine
- Department of Biomedical Engineering, McGill University
- The Research Institute of the McGill University Health Centre, Montreal, Canada
| |
Collapse
|
44
|
Bally L, Thabit H, Ruan Y, Mader JK, Kojzar H, Dellweg S, Benesch C, Hartnell S, Leelarathna L, Wilinska ME, Evans ML, Arnolds S, Pieber TR, Hovorka R. Bolusing frequency and amount impacts glucose control during hybrid closed-loop. Diabet Med 2018; 35:347-351. [PMID: 28755444 PMCID: PMC5788742 DOI: 10.1111/dme.13436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/25/2017] [Indexed: 12/16/2022]
Abstract
AIM To compare bolus insulin delivery patterns during closed-loop home studies in adults with suboptimally [HbA1c 58-86 mmol/mol (7.5%-10%)] and well-controlled [58 mmol/mol (< 7.5%)] Type 1 diabetes. METHODS Retrospective analysis of daytime and night-time insulin delivery during home use of closed-loop over 4 weeks. Daytime and night-time controller effort, defined as amount of insulin delivered by closed-loop relative to usual basal insulin delivery, and daytime bolus effort, defined as total bolus insulin delivery relative to total daytime insulin delivery were compared between both cohorts. Correlation analysis was performed between individual bolus behaviour (bolus effort and frequency) and daytime controller efforts, and proportion of time spent within and below sensor glucose target range. RESULTS Individuals with suboptimally controlled Type 1 diabetes had significantly lower bolus effort (P = 0.038) and daily bolus frequency (P < 0.001) compared with those with well-controlled diabetes. Controller effort during both daytime (P = 0.007) and night-time (P = 0.005) were significantly higher for those with suboptimally controlled Type 1 diabetes. Time when glucose was within the target range (3.9-10.0 mmol/L) during daytime correlated positively with bolus effort (r = 0.37, P = 0.016) and bolus frequency (r = 0.33, P = 0.037). Time when glucose was below the target range during daytime was comparable in both groups (P = 0.36), and did not correlate significantly with bolus effort (r = 0.28, P = 0.066) or bolus frequency (r = -0.21, P = 0.19). CONCLUSION More frequent bolusing and higher proportion of insulin delivered as bolus during hybrid closed-loop use correlated positively with time glucose was in target range. This emphasises the need for user input and educational support to benefit from this novel therapeutic modality.
Collapse
Affiliation(s)
- L. Bally
- Wellcome Trust–MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
- Department of Diabetes & EndocrinologyCambridge University Hospitals NHS Foundation TrustCambridgeUK
- Department of Diabetes & EndocrinologyClinical Nutrition and Metabolism, InselspitalBern University HospitalUniversity of BernBernSwitzerland
| | - H. Thabit
- Wellcome Trust–MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
- Department of Diabetes & EndocrinologyCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Y. Ruan
- Wellcome Trust–MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
- Department of PaediatricsUniversity of CambridgeCambridgeUK
| | - J. K. Mader
- Department of Internal MedicineDivision of Endocrinology & DiabetologyMedical University of GrazGrazAustria
| | - H. Kojzar
- Department of Internal MedicineDivision of Endocrinology & DiabetologyMedical University of GrazGrazAustria
| | - S. Dellweg
- Profil Institut fuer Stoffwechselforschung GmbHNeussGermany
| | - C. Benesch
- Profil Institut fuer Stoffwechselforschung GmbHNeussGermany
| | - S. Hartnell
- Department of Diabetes & EndocrinologyCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - L. Leelarathna
- Central Manchester University Hospitals NHS foundation Trust and University of ManchesterManchesterUK
| | - M. E. Wilinska
- Wellcome Trust–MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
- Department of PaediatricsUniversity of CambridgeCambridgeUK
| | - M. L. Evans
- Wellcome Trust–MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
- Department of Diabetes & EndocrinologyCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - S. Arnolds
- Profil Institut fuer Stoffwechselforschung GmbHNeussGermany
| | - T. R. Pieber
- Department of Internal MedicineDivision of Endocrinology & DiabetologyMedical University of GrazGrazAustria
| | - R. Hovorka
- Wellcome Trust–MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
- Department of PaediatricsUniversity of CambridgeCambridgeUK
| |
Collapse
|
45
|
Galderisi A, Sherr J, VanName M, Carria L, Zgorski M, Tichy E, Weyman K, Cengiz E, Weinzimer S, Tamborlane W. Pramlintide but Not Liraglutide Suppresses Meal-Stimulated Glucagon Responses in Type 1 Diabetes. J Clin Endocrinol Metab 2018; 103:1088-1094. [PMID: 29211871 PMCID: PMC6276715 DOI: 10.1210/jc.2017-02265] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 11/16/2017] [Indexed: 02/06/2023]
Abstract
CONTEXT Postprandial hyperglycemia remains a challenge in type 1 diabetes (T1D) due, in part, to dysregulated increases in plasma glucagon levels after meals. OBJECTIVE This study was undertaken to examine whether 3 to 4 weeks of therapy with pramlintide or liraglutide might help to blunt postprandial hyperglycemia in T1D by suppressing plasma glucagon responses to mixed-meal feedings. DESIGN Two parallel studies were conducted in which participants underwent mixed-meal tolerance tests (MMTTs) without premeal bolus insulin administration before and after 3 to 4 weeks of treatment with either pramlintide (8 participants aged 20 ± 3 years, hemoglobin A1c 6.9 ± 0.5%) or liraglutide (10 participants aged 22 ± 3 years, hemoglobin A1c 7.6 ± 0.9%). RESULTS Compared with pretreatment responses to the MMTT, treatment with pramlintide reduced the peak increment in glucagon from 32 ± 16 to 23 ± 12 pg/mL (P < 0.02). In addition, the incremental area under the plasma glucagon curve from 0 to 120 minutes dropped from 1988 ± 590 to 737 ± 577 pg/mL/min (P < 0.001), which was accompanied by a similar reduction in the meal-stimulated increase in the plasma glucose curve from 11,963 ± 1424 mg/dL/min pretreatment vs 2493 ± 1854 mg/dL/min after treatment (P < 0.01). In contrast, treatment with liraglutide had no effect on plasma glucagon and glucose responses during the MMTT. CONCLUSIONS Adjunctive treatment with pramlintide may provide an effective means to blunt postmeal hyperglycemia in T1D by suppressing dysregulated plasma glucagon responses. In contrast, plasma glucose and glucagon responses were unchanged after 3 to 4 weeks of treatment with liraglutide.
Collapse
Affiliation(s)
- Alfonso Galderisi
- Department of Pediatrics, Endocrinology and Diabetes Section, Yale School of
Medicine, New Haven, Connecticut
- Department of Women’s and Children’s Health, University of Padova, Padova,
Italy
- Correspondence and Reprint Requests: Alfonso Galderisi, MD,
Department of Pediatrics, Endocrinology and Diabetes Section, Yale School of Medicine, 333
Cedar Street, LMP3107, New Haven, Connecticut 06520. E-mail:
| | - Jennifer Sherr
- Department of Pediatrics, Endocrinology and Diabetes Section, Yale School of
Medicine, New Haven, Connecticut
| | - Michelle VanName
- Department of Pediatrics, Endocrinology and Diabetes Section, Yale School of
Medicine, New Haven, Connecticut
| | - Lori Carria
- Department of Pediatrics, Endocrinology and Diabetes Section, Yale School of
Medicine, New Haven, Connecticut
| | - Melinda Zgorski
- Department of Pediatrics, Endocrinology and Diabetes Section, Yale School of
Medicine, New Haven, Connecticut
| | - Eileen Tichy
- Department of Pediatrics, Endocrinology and Diabetes Section, Yale School of
Medicine, New Haven, Connecticut
| | - Kate Weyman
- Department of Pediatrics, Endocrinology and Diabetes Section, Yale School of
Medicine, New Haven, Connecticut
| | - Eda Cengiz
- Department of Pediatrics, Endocrinology and Diabetes Section, Yale School of
Medicine, New Haven, Connecticut
| | - Stuart Weinzimer
- Department of Pediatrics, Endocrinology and Diabetes Section, Yale School of
Medicine, New Haven, Connecticut
| | - William Tamborlane
- Department of Pediatrics, Endocrinology and Diabetes Section, Yale School of
Medicine, New Haven, Connecticut
| |
Collapse
|
46
|
|
47
|
Affiliation(s)
- David Rodbard
- Biomedical Informatics Consultants LLC , Potomac, Maryland
| |
Collapse
|
48
|
Gingras V, Taleb N, Roy-Fleming A, Legault L, Rabasa-Lhoret R. The challenges of achieving postprandial glucose control using closed-loop systems in patients with type 1 diabetes. Diabetes Obes Metab 2018; 20:245-256. [PMID: 28675686 PMCID: PMC5810921 DOI: 10.1111/dom.13052] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/27/2017] [Accepted: 06/29/2017] [Indexed: 01/17/2023]
Abstract
For patients with type 1 diabetes, closed-loop delivery systems (CLS) combining an insulin pump, a glucose sensor and a dosing algorithm allowing a dynamic hormonal infusion have been shown to improve glucose control when compared with conventional therapy. Yet, reducing glucose excursion and simplification of prandial insulin doses remain a challenge. The objective of this literature review is to examine current meal-time strategies in the context of automated delivery systems in adults and children with type 1 diabetes. Current challenges and considerations for post-meal glucose control will also be discussed. Despite promising results with meal detection, the fully automated CLS has yet failed to provide comparable glucose control to CLS with carbohydrate-matched bolus in the post-meal period. The latter strategy has been efficient in controlling post-meal glucose using different algorithms and in various settings, but at the cost of a meal carbohydrate counting burden for patients. Further improvements in meal detection algorithms or simplified meal-priming boluses may represent interesting avenues. The greatest challenges remain in regards to the pharmacokinetic and dynamic profiles of available rapid insulins as well as sensor accuracy and lag-time. New and upcoming faster acting insulins could provide important benefits. Multi-hormone CLS (eg, dual-hormone combining insulin with glucagon or pramlintide) and adjunctive therapy (eg, GLP-1 and SGLT2 inhibitors) also represent promising options. Meal glucose control with the artificial pancreas remains an important challenge for which the optimal strategy is still to be determined.
Collapse
Affiliation(s)
- Véronique Gingras
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Department of nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Nadine Taleb
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Department of biomedical sciences, Université de Montréal, Montreal, Quebec, Canada
| | - Amélie Roy-Fleming
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Department of nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Laurent Legault
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Montreal Children’s Hospital, McGill University Health Center, Montreal, Quebec, Canada
| | - Rémi Rabasa-Lhoret
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Department of nutrition, Université de Montréal, Montreal, Quebec, Canada
- Montreal Diabetes Research Center (MDRC), Montreal, Quebec, Canada
- Research Center of the Université de Montréal Hospital Center (CRCHUM), Montreal, Quebec, Canada
| |
Collapse
|
49
|
Denroche HC, Verchere CB. IAPP and type 1 diabetes: implications for immunity, metabolism and islet transplants. J Mol Endocrinol 2018; 60:R57-R75. [PMID: 29378867 DOI: 10.1530/jme-17-0138] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/06/2017] [Indexed: 01/12/2023]
Abstract
Islet amyloid polypeptide (IAPP), the main component of islet amyloid in type 2 diabetes and islet transplants, is now recognized as a contributor to beta cell dysfunction. Increasingly, evidence warrants its investigation in type 1 diabetes owing to both its immunomodulatory and metabolic actions. Autoreactive T cells to IAPP-derived epitopes have been described in humans, suggesting that IAPP is an islet autoantigen in type 1 diabetes. In addition, although aggregates of IAPP have not been implicated in type 1 diabetes, they are potent pro-inflammatory stimuli to innate immune cells, and thus, could influence autoimmunity. IAPP aggregates also occur rapidly in transplanted islets and likely contribute to islet transplant failure in type 1 diabetes through sterile inflammation. In addition, since type 1 diabetes is a disease of both insulin and IAPP deficiency, clinical trials have examined the potential benefits of IAPP replacement in type 1 diabetes with the injectable IAPP analogue, pramlintide. Pramlintide limits postprandial hyperglycemia by delaying gastric emptying and suppressing hyperglucagonemia, underlining the possible role of IAPP in postprandial glucose metabolism. Here, we review IAPP in the context of type 1 diabetes: from its potential involvement in type 1 diabetes pathogenesis, through its role in glucose metabolism and use of IAPP analogues as therapeutics, to its potential role in clinical islet transplant failure and considerations in this regard for future beta cell replacement strategies.
Collapse
Affiliation(s)
- Heather C Denroche
- Department of Surgery, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - C Bruce Verchere
- Department of Surgery, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
50
|
Otto-Buczkowska E, Jainta N. Pharmacological Treatment in Diabetes Mellitus Type 1 - Insulin and What Else? Int J Endocrinol Metab 2018; 16:e13008. [PMID: 29696037 PMCID: PMC5903388 DOI: 10.5812/ijem.13008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/18/2017] [Accepted: 10/31/2017] [Indexed: 12/19/2022] Open
Abstract
The basis of treatment in autoimmune diabetes is insulin therapy; however, many clinical cases have proven that this method does not solve all problems. Trials of causal treatment including blocking the autoimmune processes and insulin-producing cells transplants were carried out. Those methods require more research to be concerned as efficient and safe ways of treatment in type 1 diabetes. The use of non-insulin adjunct treatment is a new trend. It has been successfully used in laboratories as well as clinical trials. Metformin is the most widely used drug, together with sodium-glucose co-transporters 2 (SGLT2) inhibitors, amylin analogues, glucagon-like peptide 1 (GLP-1) receptor agonists, and dipeptidyl peptidase-4 (DPP-4) inhibitors. The results of administration of these medicaments give good outcomes in patients with diabetes mellitus type 1. Most likely, in the near future, they will progressively be used in both adult and adolescent patients with type 1 diabetes. Further multicenter, randomized studies are required to evaluate the efficacy of treatment and long term safety of these drugs.
Collapse
Affiliation(s)
- Ewa Otto-Buczkowska
- Medical Specialist Centre in Gliwice, Poland
- Corresponding author: Ewa Otto-Buczkowska MD PhD, Jasnogorska 16/2144-100 Gliwice, Poland. E-mail:
| | | |
Collapse
|