Diabetes Technology and Therapy in the Pediatric Age Group
Publication: Diabetes Technology & Therapeutics
Volume 21, Issue Number S1
Introduction
We continue to see tremendous advances in both research and clinical translation of diabetes technology and therapeutics in the pediatric age group. This year's selection of manuscripts in the article spans the full range of ages in pediatrics and includes novel glucose monitoring and closed-loop systems as well as translation of medications commonly used in adults into the pediatric population. As new technologies are researched and applied clinically, reports on use of current diabetes technologies including the effectiveness of pumps compared to multiple daily injections, use of continuous glucose monitoring (CGM) in preterm infants and data on intermittently scanned CGM (isCGM) become available. These real-world data (1) are very important for clinicians and researchers to better understand how to most effectively translate new developments to best solve the challenges our patients face on a daily basis. Further data are reported on closed-loop control systems, including predicted low-glucose suspend systems and multiple studies in which closed-loop systems were challenged by exercise, as this is a real-world problem for children and adolescents with diabetes and more data are required to best manage this in an outpatient setting.
In addition to diabetes technology, this year's articles remind us of the long-term complications of diabetes and the increased future risk of cardiovascular disease and mortality (2–4). Reports are included on therapies translated from adult to pediatric populations in studies designed to decrease the risk of long-term vascular complications. Finally, because the delay, prevention, and curing of diabetes remain the ultimate goals in diabetes research, the international TrialNet Oral Insulin trial is summarized and raises hypotheses to focus future research in type 1 diabetes (T1D).
To select these 13 articles focused on diabetes technology and therapeutics in the pediatric age group, we conducted a Medline search for articles dealing with the following topics: diabetes technology, insulin pump therapy (CSII), continuous glucose monitoring (CGM), closed-loop systems, and new therapies in T1D relating to the pediatric age group (0–18 years). We focused on key articles that offer some insight into these issues and were published between July 1, 2017, and June 30, 2018.
Insulin Pumps
Association of insulin pump therapy vs insulin injection therapy with severe hypoglycemia, ketoacidosis, and glycemic control among children, adolescents, and young adults with type 1 diabetes
Karges B1, Schwandt A2,3, Heidtmann B4, Kordonouri O5, Binder E6, Schierloh U7, Boettcher C8, Kapellen T9, Rosenbauer J10, Holl RW3
1Division of Endocrinology and Diabetes, Medical Faculty, RWTH Aachen University, Aachen, Germany; 2Institute of Epidemiology and Medical Biometry, ZIBMT, University of Ulm, Ulm, Germany; 3German Center for Diabetes Research (DZD), Neuherberg, Germany; 4Children's Hospital Wilhelmstift, Hamburg, Germany; 5Diabetes Center for Children and Adolescents, Children's Hospital Auf Der Bult, Hannover, Germany; 6Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria; 7Department of Pediatrics, Centre Hospitalier de Luxembourg, Luxembourg City, Luxembourg; 8Division of Paediatric Endocrinology and Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University Giessen, Giessen, Germany; 9Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany; 10Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center at the University of Düsseldorf, Düsseldorf, Germany
This manuscript is also discussed in the article on Insulin Pumps, page S-32.
Background
The use of insulin pumps for intensive insulin therapy among patients with type 1 diabetes (T1D) has substantially increased. The aim of this study was to determine whether rates of short-term diabetes complications, namely severe hypoglycemia and diabetic ketoacidosis (DKA), are lower with insulin pump therapy compared with insulin injection therapy in children, adolescents, and young adults with T1D.
Methods
A population-based cohort study was conducted between January 2011 and December 2015 in 446 diabetes centers participating in the Diabetes Prospective Follow-up Initiative in Germany, Austria, and Luxembourg. Study participants were patients younger than 20 years of age with T1D and diabetes duration >1 year, treated with insulin pump therapy or with multiple daily insulin injections. The main outcomes were rates of severe hypoglycemia and DKA during the most recent treatment year. Secondary outcomes included glycated hemoglobin levels, insulin dose, and body mass index.
Results
Of 30,579 patients (53% male) with a mean age of 14.1±4.0 years, 14,119 (46.2%) used pump therapy (median duration, 3.7 years) and 16,460 used insulin injections (median duration, 3.6 years). Patients using pump therapy (n=9814) were matched with patients using injection therapy (n=9814). Compared with injection therapy, pump therapy was associated with lower rates of both severe hypoglycemia (9.55 vs 13.97 per 100 patient-years; P<0.001) and DKA (3.64 vs 4.26 per 100 patient-years; P=0.04). Glycated hemoglobin levels and total daily insulin doses were also lower with pump therapy than with injection therapy (8.04% vs 8.22%; P<0.001 and 0.84U/kg vs 0.98U/kg; P<0.001, respectively). No significant difference was observed for body mass index between the two treatment regimens.
Conclusions
Among young patients with T1D, insulin pump therapy was associated with improved clinical outcomes determined by reduced risks of short-term diabetes complications including severe hypoglycemia and DKA as well as better glycemic control during the most recent year of therapy compared with insulin injection therapy.
Blood Glucose Monitoring
Continuous glucose monitoring in very preterm infants: a randomized controlled trial
Galderisi A1,2, Facchinetti A3, Steil GM4, Ortiz-Rubio P4, Cavallin F5, Tamborlane WV2, Baraldi E1, Cobelli C3, Trevisanuto D1
1NICU, Departments of Women's and Child's Health and 2Endocrinology Section, Department of Pediatrics, Yale University, New Haven, CT; 3Information Engineering, University of Padova, Padova, Italy; 4Boston Children's Hospital, Boston, MA; 5Independent Statistician, Padova, Italy
Background
Very preterm infants (≤32 weeks gestation or birth weight ≤1500 g) are at risk for morbidity, mortality, and neurologic consequences from abnormal blood sugar. The aim of this study was to assess whether glucose administration guided by continuous glucose monitoring (CGM) was more effective than standard of care in maintaining euglycemia in very preterm infants.
Methods
Within the first 48 hours of life, 50 very preterm infants were randomly assigned to receive computer-guided intravenous dextrose driven by CGM or standard of care glucometer. The primary outcome was time spent with blood sugar of 72–144 mg/dL. Secondary outcomes included time with blood sugar <47mg/dL (severe hypoglycemia), 47–71 mg/dL (mild hypoglycemia), 145–180 mg/dL (mild hyperglycemia), >180 mg/dL (severe hyperglycemia), glucose variability, and clinical outcomes.
Results
Those in the group using CGM-mediated dextrose delivery spent 84% time in range vs 68% for the standard of care group (P<0.001). There was a statistically significant reduction in time spent in severe hypoglycemia (P=0.007), mild hypoglycemia (P=0.04), and severe hyperglycemia (P=0.04) with reduction in glucose variability (mean ± SD 21.6±5.4 mg/dL vs 27±7.2 mg/dL, P=0.01; coefficient of variation 22.8% ±4.2% vs 27.9% ±5.0%, P<0.001).
Conclusions
CGM-guided titration of intravenous dextrose can successfully increase time in range, reduce hypoglycemia, and minimize glycemic variability in preterm infants during the first week of life.
Effect of financial incentives on glucose monitoring adherence and glycemic control among adolescents and young adults with type 1 diabetes: a randomized clinical trial
Wong CA1,2, Miller VA3, Murphy K4, Small D2,5, Ford CA3, Willi SM6, Feingold J7, Morris A8, Ha YP8, Zhu J2,8, Wang W2,8, Patel MS2,8,9,10
1Department of Pediatrics, Duke Clinical Research Institute, Duke-Margolis Center for Health Policy, Duke University, Durham, NC; 2Leonard Davis Institute of Health Economics, Center for Health Incentives and Behavioral Economics at the University of Pennsylvania, Philadelphia, PA; 3Division of Adolescent Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine and University of Pennsylvania, Philadelphia, PA; 4Division of Pediatric Endocrinology, The Children's Hospital of Philadelphia, Philadelphia, PA; 5Department of Statistics, The Wharton School, University of Pennsylvania, Philadelphia, PA; 6Division of Pediatric Endocrinology, The Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; 7Medical student, Icahn School of Medicine at Mount Sinai, New York, NY; 8Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; 9Department of Medicine, Crescenz Veterans Affairs Medical Center, Philadelphia, PA; 10Health Care Management, The Wharton School, University of Pennsylvania, Philadelphia, PA
This manuscript is also discussed in the article on Self-Monitoring of Blood Glucose, page S-4.
Background
For those with type 1 diabetes, glucose control often worsens during the transition to young adulthood, which may lead to complications later in life. The aim of this study was to determine the effect of financial incentives on adherence to glucose checks and the effect on glycemic control in adolescents and young adults with type 1 diabetes (T1D).
Methods
The BE IN CONTROL (Behavioral Economic Incentives to Improve Glycemic Control Among Adolescents and Young Adults with T1D) study was a 6-month, two-arm, randomized clinical trial with 3-month intervention and follow-up periods. Ninety participants with T1D and HbA1c >8.0% were randomized to receive either standard of care for 6 months or 3 months of an incentive program followed by 3 months standard of care. The daily blood glucose monitoring goal was four or more checks per day with one or more readings of 70–180 mg/dL. The incentive was $60/month, from which $2 was subtracted for every day goals were not met. During the following 3 months, the incentive was discontinued. The primary outcome was change in HbA1c at 3 months. Secondary outcomes included frequency of glucose monitoring and change in HbA1c at 6 months.
Results
During the first 3-month period, daily adherence to glucose checks was significantly greater (P=0.003) in the incentive group (50.0%) vs the control group (18.9%). The difference was not maintained after the incentive was removed over the subsequent 3 months. HbA1c did not significantly differ between groups at 3 or 6 months.
Conclusions
Adolescents and young adults with T1D displayed improved glucose monitoring adherence during a 3-month daily financial incentive. This did not translate into significantly improved glycemic control.
Flash glucose measurements in children with type 1 diabetes in real-life settings: to trust or not to trust?
Szadkowska A1, Gawrecki A2, Michalak A1, Zozulińska-Ziółkiewicz D2, Wojciech Fendler W1,3, Młynarski W1
1Departments of Pediatrics, Oncology, Hematology and Diabetology and 3Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland; 2Department of Internal Medicine and Diabetology, Poznan University of Medical Science, Poznan, Poland
Background
Maintaining a high frequency of self-monitoring of blood glucose (SMBG) is the cornerstone for meeting HbA1c goals, preventing serious hypoglycemia, and associated with significantly lower rates of diabetic ketoacidosis. CGM offers efficient glycemic control with a reduced need for manual tests and associated inconveniences. A new intermittently scanned CGM (isCGM) system (Abbott Diabetes Care, Alameda, CA), FreeStyle Libre (FSL), was proposed as an alternative. The main aim of the study was to establish the clinical accuracy of a Flash glucose-monitoring device FSL in reference to the glucometer in children with type 1 diabetes (T1D) in a real-world setting during a summer camp. The secondary objective was to assess the children's attitude toward the device and its possible association with measurement accuracy.
Methods
This was a prospective, single-arm study conducted during an annual summer camp for children and adolescents with T1D. During the camp, children (n=79, 8–18 years of age) were provided with FSLs for 12 days. Supervised glucose testing was carried out at eight time points on days 3, 7, and 11 of the study. Glycemia was estimated using FSL and measured with a personal glucometer within 2 minutes, and glucose trend arrows were recorded. On the last day, a custom-designed questionnaire was administered to assess the degree of satisfaction with isCGM use.
Results
The study participants (n=78; 43% boys) had a median age of 12.8 years (interquartile range [IQR] 11.6–14.7 years), with a median diabetes duration of 5.8 years (IQR 3.8–8.5 years). Their median HbA1c at inclusion was 7.5% (IQR 7.0%–8.0%) or 58.5 mmol/mol (IQR 53–63.9 mmol/mol); 88.6% were treated with insulin pump therapy. During the summer camp, children wore the sensors for a median time of 10.5 days (IQR 10 to complete 11 days), and 10 sensors were replaced in eight children due to detachment. Mean absolute relative difference (MARD) between the FSL and the glucometer was 13.5%–12.9%. FSL was the most accurate in stable glycemic conditions (MARD 11.4%–10.4%), less accurate when glycemia was falling >2 mg/(dL. min) [0.111 mmol/(L. min): MARD 22.6%–18.6%; P<0.001 vs stable conditions], and when the device could not determine the glucose trends (16.5%–16.3%; P=0.01 vs stable conditions). The FSL was less accurate during the day than during the night (MARD 14.9%–14% vs 11.2%–10.6%; P<0.0001). The Surveillance Error Grid methodology was used to evaluate paired results: 80.36% of FSL readings were associated with no clinical risk, 18.73% were associated with slight risk, and one high-risk measurement was detected. Sixty-eight (87.2%) children expressed a willingness to continue isCGM use. Most of the participants regarded the discretion and painlessness of measurements as key advantages and its inaccuracy, as perceived by the children, as a disadvantage.
Conclusions
The FSL isCGM device offers good accuracy in comparison with capillary blood glucose in children and adolescents with T1D. However, the performance of isCGM is strongly affected by the glycemia change trend at the time of measurement. Therapeutic decisions should not be solely based on measurements flagged by a rapid change flag on the FSL isCGM. Such measurements should first be verified by using a blood glucose measurement.
Reduction in hypoglycemia with the predictive low-glucose management system: a long-term randomized controlled trial in adolescents with type 1 diabetes
Abraham MB1,3, Nicholas JA1,2, Smith GJ2, Fairchild JM4, King BR5, Ambler GR6, Cameron FJ7, Davis EA1,3 Jones TW1,3; on behalf of the PLGM Study Group
1Children's Diabetes Centre, Telethon Kids Institute, The University of Western Australia, Perth, Australia; 2Department of Endocrinology and Diabetes, Princess Margaret Hospital for Children, Perth, Australia; 3Division of Paediatrics, Medical School, The University of Western Australia, Perth, Australia; 4Department of Endocrinology and Diabetes, Women's and Children's Hospital, Adelaide, Australia; 5Department of Endocrinology and Diabetes, John Hunter Children's Hospital, Newcastle, Australia; 6Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead and Clinical School, The University of Sydney, Sydney, Australia; 7Department of Endocrinology and Diabetes, The Royal Children's Hospital, Melbourne, Australia
This manuscript is also discussed in the article on Insulin Pumps, page S-32.
Background
Short-term studies with automated systems that suspend basal insulin for predicted hypoglycemia reduce hypoglycemia, but long-term efficacy and safety have not been established.
Methods
A 6-month, multicenter, randomized controlled trial was carried out in children and adolescents with T1D, comparing the use of Medtronic MiniMed 640G pump with predictive low-glucose management (PLGM) with sensor-augmented pump therapy (SAPT) alone. The percentage time in hypoglycemia with sensor glucose (SG) <3.5 mmol/L (63 mg/dL) was the primary outcome.
Results
Seventy-four subjects were randomized to SAPT and 80 subjects to PLGM in an intent-to-treat analysis. The time with SG <3.5 mmol/L was 3.0% in the SAPT group and 2.8% in the PLGM group at baseline. PLGM was associated with a reduction in hypoglycemia compared with SAPT (% time SG <3.5 mmol/L SAPT vs PLGM, 2.6 vs 1.5; P<0.0001), as was time with SG <3 mmol/L (P<0.0001). This reduction was seen during both day and night (P<0.0001). Hypoglycemic events (SG <3.5 mmol/L for >20 min) also declined with PLGM vs SAPT (139 vs 227 events/patient-year; P<0.001). There was no difference in HbA1c at 6 months (SAPT 7.6±1.0% vs PLGM 7.8±0.8%; P=0.35). No PLGM-related serious adverse events were reported, and quality of life measures in did not change in either the parent group or the participant group.
Conclusions
PLGM reduced hypoglycemia without deterioration in HbA1c in children and adolescents with T1D.
Closed-Loop Insulin Delivery Systems
Closed-loop control during intense prolonged outdoor exercise in adolescents with type 1 diabetes: the artificial pancreas ski study
Breton MD1, Cherñavvsky DR1, Forlenza GP2, DeBoer MD1, Robic J2, Wadwa RP2, Messer LH2, Kovatchev BP1, Maahs DM2,3
1Center for Diabetes Technology, University of Virginia, Charlottesville, VA; 2Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO; 3Department of Pediatrics, Stanford University, Stanford, CA
This manuscript is also discussed in the article on Decision Support Systems and Closed Loop, page S-42, and the article on Advances in Exercise, Physical Activity, and Diabetes Mellitus, page S-112.
Background
Intense exercise challenges type 1 diabetes (T1D) management. Closed-loop control (CLC) systems improve glycemic control during physical activity of limited intensity and short duration. However, CLC data are limited for extended vigorous outdoor exercise among adolescents.
Methods
Skiing presents unique metabolic challenges: intense prolonged exercise, cold, altitude, and stress/fear/excitement. Thirty-two adolescents with T1D (10–16 years of age) participated in a randomized controlled trial during two 5-day ski camps (5 h skiing/∼day) in Virginia and Colorado. Participants were randomly assigned to the University of Virginia CLC system or remotely monitored sensor-augmented pump (RM-SAP). The CLC and RM-SAP groups were paired by age and hemoglobin A1c (HbA1c) and remotely monitored by the study team 24 h/day.
Results
Percent time in range (70–180 mg/dL) improved using CLC: 71.3 vs 64.7% (+6.6% [95% confidence interval 1–12]; P=0.005) compared with physician-monitored open loop. Maximum effect was reported late at night. Both hypoglycemia exposure and carbohydrate treatments improved overall (P=0.001 and P=0.007) and during the daytime, although ski level effects were strong (P=0.0001 and P=0.006). The two groups were balanced in terms of ski/snowboard proficiency, but there was a very strong effect by site (experienced in Colorado and naive in Virginia). No adverse events were reported with CLC, and feedback from participants was strongly positive.
Conclusions
CLC in adolescents with T1D reduced hypoglycemia and improved glycemic control during prolonged intensive winter sports, including challenges of altitude and cold.
Optimizing hybrid closed-loop therapy in adolescents and emerging adults using the MiniMed 670G system
Messer LH1, Forlenza GP1, Sherr JL2, Wadwa RP1, Buckingham BA3, Weinzimer SA2, Maahs DM3, Slover RH1
1Barbara Davis Center for Childhood Diabetes, University of Colorado, Anschutz Medical Campus, Aurora, CO; 2Department of Pediatrics, Yale School of Medicine, New Haven, CT; 3Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
This manuscript is also discussed in the article on Diabetes Technology and the Human Factor, page S-138.
Background
The MiniMed 670G System is the first globally available commercial hybrid closed-loop (HCL) system for management of type 1 diabetes (T1D). Insulin delivery patterns and time-in-range metrics were investigated in HCL “Auto Mode” and open-loop (OL) “Manual Mode” in adolescents and young adults with T1D. We examined system alerts, usage profiles, and operational parameters with the objective of providing guidance for optimal clinical use of the system.
Methods
We analyzed data from 31 adolescent and young adult participants (14–26 years old) at three clinical sites in the 670G pivotal trial. Participants completed a 2-week run-in period in OL (insulin pump with CGM), followed by a 3-month in-home study phase with HCL functionality (Auto Mode) enabled. Data were compared between baseline OL and HCL use after 1 week, 1 month, 2 months, and 3 months.
Results
Compared with baseline OL, carbohydrate-to-insulin (C-to-I) ratios were more aggressive for all meals with HCL, although total daily insulin dose and ratio of basal to bolus was unchanged over the duration of the study. HbA1c decreased 0.75% and time in range increased 14% with use of Auto Mode after 3 months (P<0.001). Time in Auto Mode declined from 87% to a final use rate of 72% (−15%). The main causes of Auto Mode exit were sensor/insulin delivery alerts and hyperglycemia.
Conclusions
For adolescents and young adults who transition to the MiniMed 670G System, health-care providers should anticipate immediate C-to-I ratio adjustments and assess active insulin time. Users should anticipate occasional Auto Mode exits and should follow system instructions and bolus for meals to reduce exits. Ongoing clinical guidance and education from providers will be required for 670G optimization.
Closed-loop glucose control in young people with type 1 diabetes during and after unannounced physical activity: a randomised controlled crossover trial
Dovc K1, Macedoni M2, Bratina N1, Lepej D3, Nimri R4, Atlas E5, Muller I5, Kordonouri O6, Biester T6, Danne T6, Phillip M4,7, Battelino T8,9
1Department of Paediatric Endocrinology, Diabetes and Metabolic Diseases, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia; 2Department of Paediatrics – Diabetes Service Studies, University of Milan, Ospedale dei Bambini Vittore Buzzi, Milan, Italy; 3Department of Pulmonology, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia; 4The Jesse and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Centre for Childhood Diabetes, Schneider Children's Medical Centre of Israel, Petah Tikva, Israel; 5DreaMed Diabetes Ltd, Petah Tikva, Israel; 6Diabetes Centre for Children and Adolescents, Kinder- und Jugendkrankenhaus Auf der Bult, Hannover, Germany; 7Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; 8Department of Paediatric Endocrinology, Diabetes and Metabolic Diseases, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia; 9Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
This manuscript is also discussed in the article on Decision Support Systems and Closed Loop, page S-42, and the article on Advances in Exercise, Physical Activity, and Diabetes Mellitus, page S-112.
Background
Hypoglycemia associated with exercise remains a challenge for patients with T1D. This study tested the safety and performance of the Glucositter fuzzy-logic controller on young people with T1D engaging in a regimented exercise routine conducted in the hospital.
Methods
During four visits at a single site in Slovenia, individuals with T1D experienced with pump use were randomized to perform two separate exercise routines with closed- or open-loop insulin delivery. The participants performed unannounced exercise that was either moderate intensity (55% of VO2 max) or moderate intensity with integrated high-intensity sprints (55/80% of VO2 max). Closed-loop insulin delivery occurred from 3 p.m. on the day of exercise to 1 p.m. the next day. The primary outcome was glucose control during the exercise period and the following night.
Results
Twenty participants with T1D performed all activities. Neither group experienced hypoglycemia below 60mg/dL. However, closed-loop insulin delivery increased time in range to 84.1% vs 68.7% for the open-loop group (P=0.0057) with a decrease in required insulin (P=0.0123).
Conclusions
Closed-loop insulin delivery resulted in no hypoglycemia during this in-hospital trial. There was a significant increase in time in range for closed-loop versus open-loop insulin delivery.
Safety and feasibility of the OmniPod hybrid closed-loop system in adult, adolescent, and pediatric patients with type 1 diabetes using a personalized model predictive control algorithm
Buckingham BA1, Forlenza GP2, Pinsker JE3, Christiansen MP4, Wadwa RP2, Schneider J5, Peyser TA5, Dassau E6, Lee JB7, O'Connor J7, Layne JE7, Ly TT7
1Division of Endocrinology, Department of Pediatrics, Stanford School of Medicine, Stanford, Palo Alto, CA; 2Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO; 3Sansum Diabetes Research Institute, Santa Barbara, CA; 4Diablo Clinical Research, Walnut Creek, CA; 5ModeAGC LLC, Palo Alto, CA; 6Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA; 7Insulet Corporation, Billerica, MA
Background
The current study assesses the Insulet model predictive control algorithm in youth, adolescents, and adults with type 1 diabetes (T1D). Insulet's system utilizes the OmniPod patch pump, Dexcom G4 CGM, and a model predictive control algorithm running on a tablet for automated insulin delivery.
Methods
This was a multicenter safety and feasibility trial with 1-week use of only sensor and pump followed by 36 hours on the closed-loop system. Subjects with T1D who were 6–65 years of age with hemoglobin A1c level of 6%–10% were eligible. Physical activity was limited, and announced meals ranged from 30 to 90 grams of carbohydrates. Primary endpoints included percent time with sensor glucose <70 mg/dL and >250 mg/dL.
Results
Adults receiving 80% bolus had 0.7% time <70 mg/dL, 69.5% time 70–180 mg/dL, and 8% time >250 mg/dL; adults receiving full bolus 0.7% time <70 mg/dL, 73.0% time 70–180 mg/dL, and 3.6% time >250 mg/dL; adolescents 2% time <70 mg/dL, 72.6% time 70–180 mg/dL, and 4.9% time >250 mg/dL; and pediatric patients 2% time <70 mg/dL, 70.1% time 70–180 mg/dL, and 6.7% time >250 mg/dL.
Conclusions
The authors conclude that the device is safe for further testing in those with T1D.
Other Treatments for Diabetes
ACE inhibitors and statins in adolescents with type 1 diabetes
Marcovecchio ML1, Chiesa ST4, Bond S3, Daneman D8, Dawson S3, Donaghue KC9, Jones TW10, Mahmud FH8, Marshall SM6, Neil HAW7, Dalton RN5, Deanfield J4, Dunger DB1,2; for the AdDIT Study Group
1The Department of Paediatrics and 2the Wellcome Trust–Medical Research Council Institute of Metabolic Science, University of Cambridge, UK; 3The Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, UK; 4Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London, UK; 5The Well Child Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital, London, UK; 6The Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; 7The Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK; 8The Department of Paediatrics, Hospital for Sick Children, and University of Toronto, Toronto, Australia; 9The Institute of Endocrinology and Diabetes, Children's Hospital at Westmead, and University of Sydney, Sydney, Australia; 10The Telethon Kids Institute, University of Western Australia, Perth, Australia
Background
Rapid increases in the excretion of albumin during puberty precede the development of microalbuminuria and macroalbuminuria in adolescent patients with type 1 diabetes (T1D). These changes are associated with dyslipidemia, hypertension, increased levels of high-sensitivity C-reactive protein, increased aortic and carotid intima-media thickness, and altered retinal vasculature. The aim was to evaluate whether adolescents with high levels of albumin excretion might benefit from therapy with angiotensin-converting enzyme (ACE) inhibitors and statins.
Methods
The trial was a double-blind, randomized, placebo-controlled trial at 32 centers in three countries (the United Kingdom, Canada, and Australia) designed to explore the effects of two medications with different mechanisms of action in a population of adolescents with T1D. Adolescents aged 10–16 years were screened (n=4407), and 1287 were identified with values in the upper third of the albumin/creatinine ratios. Patients (n=443) underwent randomization to one of four regimens: ACE inhibitor plus placebo, statin plus placebo, ACE inhibitor plus statin, or placebo plus placebo. The primary outcome for both interventions was the change in albumin excretion, assessed by albumin/creatinine ratio calculated from three early-morning urine samples obtained every 6 months over 2–4 years, and expressed as the area under the curve. Secondary outcomes included the development of microalbuminuria, progression of retinopathy, changes in the glomerular filtration rate, lipid levels, and measures of cardiovascular risk assessed by the carotid intima–media thickness, and levels of high-sensitivity C-reactive protein and asymmetric dimethylarginine.
Results
Overall adherence to the drug regimen was 75%, and there were no significant differences in the rate of serious adverse events between groups. The change in albumin excretion was not affected by ACE inhibitor therapy, statin therapy, or the combination of the two. The use of an ACE inhibitor was associated with a lower incidence of microalbuminuria than was the use of placebo, although it was not statistically significant. Significant reductions were observed in total, low-density lipoprotein, and non–high-density lipoprotein cholesterol levels and triglyceride levels, as well as in the ratio of apolipoprotein B to apolipoprotein A1, with the use of statins. However, neither medication caused significant changes in carotid intima-media thickness, other cardiovascular markers, the glomerular filtration rate, or the progression of retinopathy.
Conclusions
Neither ACE inhibitor nor statin therapy over a period of 2–4 years altered the primary outcome of the change in repeated measures of the albumin-to-creatinine ratio among adolescents with T1D.
Effect of metformin on vascular function in children with type 1 diabetes: a 12-month randomized controlled trial
Anderson JJA1,2, Couper JJ1,2, Giles LC3, Leggett CE1,4, Gent R5, Coppin B6, Peña AS1,2
1Discipline of Paediatrics, Robinson Research Institute, University of Adelaide, North Adelaide, South Australia, Australia; 2Endocrinology and Diabetes Department, Women's and Children's Hospital, North Adelaide, South Australia, Australia; 3School of Public Health, Faculty of Health and Medical Sciences, University of Adelaide, North Adelaide, South Australia, Australia; 4Pharmacy, Women's and Children's Hospital, North Adelaide, South Australia, Australia; 5Medical Imaging, Women's and Children's Hospital, North Adelaide, South Australia, Australia; 6Flinders Medical Centre, Bedford Park, South Australia, Australia
Background
Patients with T1D have three times the risk for cardiovascular causes of death as matched controls, despite good glycemic control. Therefore, they need additional strategies to improve cardiovascular health. Vascular dysfunction is a critical event in the development of cardiovascular disease (CVD) and is detectable years before CVD develops. Vascular function is impaired in children with T1D, and the early vascular changes in function and structure are potentially reversible. The aim of the study was to evaluate the effect of metformin on vascular health in children with T1D and with a weight above the average.
Methods
In this 12-month double-blind, randomized, placebo-controlled trial, children (n=90) aged 8–18 years with T1D of at least 6-month duration, and with a body mass index (BMI) >50th percentile for age and sex were randomly allocated in a 1:1 ratio to metformin (up to 1 g twice a day) or placebo. The main outcome was the vascular function measured by brachial artery ultrasound (flow-mediated dilatation/glyceryl trinitrate–mediated dilatation [GTN]). Other outcomes included HbA1c, insulin dose, BMI, body composition, waist circumference, mean of three consecutive blood pressures, fasting lipid profile, high-sensitivity C-reactive protein, adiponectin, leptin, and early morning urinary albumin/creatinine ratio.
Results
Ninety participants (41 boys) were enrolled. Mean age of participants was 13.6±3.5 years. Median (interquartile range) HbA1c was 8.7% (8.1–9.9) (72 mmol/mol [65–85]) and mean BMI z-score was 0.89±0.57. Fifty-four participants had normal BMI (BMI 50%–84%), 25 were overweight (BMI 85%–95%), and 11 were obese (BMI >95%). Ten patients discontinued intervention, and one was lost to follow-up. Median (95% confidence interval [CI]) adherence, evaluated by electronic monitoring, was 75.5% (65.7–81.5), without group differences. On metformin, GTN improved, independent of HbA1c, by 3.3 percentage units ([95% CI 0.3–6.3]; P=0.03) and insulin dose was reduced by 0.2 U/kg/d ([95% CI 0.1–0.3] P=0.001) during 12 months, with effects from 3 months. There was a significant benefit in adjusted (age, sex) HbA1c at 3 months for the metformin group (8.4% [95% CI 8.0–8.8]) (68 mmol/mol [95% CI 64–73]) vs placebo group (9.3% [95% CI 9.0–9.7]) (78 mmol/mol [95% CI 75–83]) (P=0.001), and this was primarily responsible for the overall benefit of metformin compared with placebo during the study period. There were no effects on carotid/aortic intima-media thickness, BMI, lipids, blood pressure, or other cardiovascular risk factors. Gastrointestinal side effects were more common in the metformin group compared with placebo (incidence rate ratio 1.65 [95% CI 1.08–2.52]; P=0.02), with no difference in hypoglycemia or diabetic ketoacidosis.
Conclusions
The study findings demonstrated the benefit of metformin on vascular smooth muscle function, HbA1c, insulin dose, and estimated insulin sensitivity during 12 months in children with T1D who are above the average weight as well as the drug's good safety profile.
Randomized, double-blind, placebo-controlled dose-finding study of the dipeptidyl peptidase-4 inhibitor linagliptin in pediatric patients with type 2 diabetes
Tamborlane WV1, Laffel LM2, Weill J3, Gordat M4, Neubacher D5, Retlich S5, Hettema W5, Hoesl CE5, Kaspers S6, Marquard J6
1Department of Pediatrics, Yale School of Medicine, New Haven, CT; 2Joslin Diabetes Center, Harvard Medical School, Boston, MA; 3Hôpital Jeanne de Flandre, Lille, France; 4Boehringer Ingelheim, Reims, France; 5Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany; 6Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
Background
The limited treatment options currently available for youth with T2D stand in contrast to the number of new medications (e.g., dipeptidyl peptidase-4 [DPP-4] inhibitors, glucagon-like peptide-1 [GLP-1] agonists, and sodium glucose cotransporter 2 [SGLT2] inhibitors) that have been approved for use in adults with T2D. Linagliptin is a potent DPP-4 inhibitor and is eliminated predominantly by nonrenal excretion and can be prescribed to adult patients with T2D without adjusting the dose according to kidney function. The aim of this study was to provide the pharmacokinetics (PK)/pharmacodynamics (PD) data to inform the choice of the appropriate dose of linagliptin to be used in the pediatric patients with T2D.
Methods
Thirty-nine patients 10–18 years of age with T2D were included in a double-blind, randomized, placebo-controlled study that compared linagliptin 1 mg and 5 mg once daily (approved for use in adults with T2D) with placebo. The primary efficacy endpoint was the change from baseline in HbA1c after 12 weeks of treatment. The key pharmacodynamic endpoint was DPP-4 inhibition during steady-state.
Results
Compared with placebo, there was a dose-dependent reduction in mean HbA1c of 0.48% and 0.63% with linagliptin 1 mg and 5 mg, respectively, with corresponding decreases in mean fasting plasma glucose of 5.6 and 34.2 mg/dL. Median DPP-4 inhibition was 38% with linagliptin 1 mg and 79% with linagliptin 5 mg. Geometric mean trough levels of linagliptin were 3.80 nmol/L and 7.42 nmol/L in the 1 mg and 5 mg groups, respectively. The levels were slightly higher than those in adult patients with T2D, probably due to higher plasma DPP-4 concentrations in the study population. There were no drug-related adverse events during treatment with either dose of linagliptin.
Conclusions
Overall, the results are consistent with the clinical efficacy and safety profile that has been reported for linagliptin in adult patients with T2D, favoring linagliptin 5 mg over 1 mg. Linagliptin was well tolerated and induced dose-dependent DPP-4 inhibition that was accompanied by corresponding reductions in HbA1c and fasting plasma glucose levels in children with T2D.
Effect of oral insulin on prevention of diabetes in relatives of patients with type 1 diabetes: a randomized clinical trial
Writing Committee for the Type 1 Diabetes TrialNet Oral Insulin Study Group, Krischer JP1, Schatz DA2, Bundy B1, Skyler JS3, Greenbaum CJ4
1University of South Florida, Tampa, FL; 2University of Florida, Gainesville, FL; 3University of Miami, Miami, FL; 4Benaroya Research Institute, Seattle, WA
This manuscript is also discussed in the article on Immune Intervention in Type 1 Diabetes, page S-95.
Background
Patients with type 1 diabetes (T1D) face major lifestyle changes and increased morbidity and mortality. Therefore, preventing or delaying the onset of diabetes would provide major public health benefit. The objective of this study was to determine whether oral insulin delays onset of T1D in autoantibody-positive relatives of patients with T1D.
Methods
Between March 2, 2007, and December 21, 2015, relatives with at least two autoantibodies, including insulin autoantibodies and normal glucose tolerance, were enrolled in nine countries worldwide. The main study group (n=389) had first-phase insulin release on an intravenous glucose tolerance test that was higher than the threshold. The secondary stratum 1 (n=55) included individuals with an antibody profile identical to the main study group but who had first-phase insulin release that was lower than the threshold. Secondary strata 2 (n=114) and strata 3 (n=3) had different autoantibody profiles and first-phase insulin release threshold combinations. Participants were randomized to receive 7.5 mg/day of oral insulin (n=283) or placebo (n=277). The primary outcome was time to diabetes in the main study group. A one-sided threshold of 0.05 determined significance, and one-sided 95% confidence intervals (95% CI) are reported.
Results
A total of 560 randomized participants were enrolled in the trial (median enrollment age, 8.2 years; interquartile range [IQR] 5.7–12.1 years; 60% male, n=170; 90.7% white non-Hispanic; 57.6% with a sibling with T1D). Of those enrolled, 550 participants completed the trial, including 389 participants (median age 8.4 years; 245 boys [63%]) in the main study group. T1D was diagnosed in 58 subjects (28.5%) in the oral insulin group and 62 (33%) in the placebo group during a median follow-up of 2.7 years (IQR 1.5–4.6 years) in the main study group. There was no significant difference in time to T1D between the oral insulin and placebo groups (hazard ratio 0.87 [95% CI 0–1.2]; P=0.21). In secondary stratum 1 (n=55), T1D was diagnosed in 13 participants (48.1%) in the oral insulin group and 19 participants (70.3%) in the placebo group, with a significantly longer time to T1D with oral insulin (HR 0.45 [95% CI 0–0.82]; P=0.006). For between-group comparisons among the 116 participants in the other secondary strata, the hazard ratio for time to T1D was 1.03 ([95% CI 0–2.11]; P=0.53), and the hazard ratio for the entire cohort of 560 participants was 0.83 ([95% CI 0–1.07]; P=0.11), which were not significantly different. With 254 events, infection was the most common adverse event (n=134 for oral insulin group; n=120 for placebo), but no significant study-related adverse events occurred.
Conclusions
In this trial of oral insulin vs placebo among autoantibody-positive relatives of patients with T1D, 7.5 mg/day of oral insulin did not delay or prevent the development of T1D over 2.7 years. These findings do not support oral insulin as used in this study for T1D prevention.
References
1. Jarow JP, LaVange L, Woodcock J. Multidimensional evidence generation and FDA regulatory decision making: defining and using “real-world” data. JAMA 2017; 318: 703–704.
2. Bjornstad P, Donaghue KC, Maahs DM. Macrovascular disease and risk factors in youth with type 1 diabetes: time to be more attentive to treatment? Lancet Diabetes Endocrinol 2018; 6: 809–820.
3. Rawshani A, Sattar N, Franzén S, et al. Excess mortality and cardiovascular disease in type 1 diabetes in relation to age at onset: a nationwide study of 27,195 young adults with diabetes. Lancet 2018; 392: 477–486.
4. Basina M, Maahs DM. Age at type 1 diabetes onset: a new cardiovascular disease risk factor and call for focused treatment? Lancet 2018; 392: 453–454.
5. Sherr JL, Hermann JM, Campbell F, et al; T1D Exchange Clinic Network, the DPV Initiative, and the National Paediatric Diabetes Audit and the Royal College of Paediatrics and Child Health registries. Use of insulin pump therapy in children and adolescents with type 1 diabetes and its impact on metabolic control: comparison of results from three large, transatlantic paediatric registries. Diabetologia 2016; 59: 87–91.
6. Szypowska A, Schwandt A, Svensson J, et al; SWEET Study Group. Insulin pump therapy in children with type 1 diabetes: analysis of data from the SWEET registry. Pediatr Diabetes 2016; 17(suppl 23): 38–45.
7. Blackman SM, Raghinaru D, Adi S, et al. Insulin pump use in young children in the T1D Exchange clinic registry is associated with lower hemoglobin A1c levels than injection therapy. Pediatr Diabetes 2014; 15: 564–572.
8. Brorsson AL, Viklund G, Örtqvist E, Lindholm Olinder A. Does treatment with an insulin pump improve glycaemic control in children and adolescents with type 1 diabetes? a retrospective case-control study. Pediatr Diabetes 2015; 16: 546–553.
9. Hanas R, Lindgren F, Lindblad B. A 2-yr national population study of pediatric ketoacidosis in Sweden: predisposing conditions and insulin pump use. Pediatr Diabetes 2009; 10: 33–37.
10. McKinlay CJD, Alsweiler JM, Anstice NS, et al. Children with hypoglycemia and their later development (CHYLD) study team. “Association of neonatal glycemia with neurodevelopmental outcomes at 4.5 years.” JAMA Pediatr 2017; 171: 972–983.
11. Miller KM, Foster NC, Beck RW, et al; T1D Exchange Clinic Network. Current state of type 1 diabetes treatment in the US: updated data from the T1D Exchange Clinic registry. Diabetes Care 2015; 38: 971–978.
12. Borus JS, Laffel L. Adherence challenges in the management of type 1 diabetes in adolescents: prevention and intervention. Curr Opin Pediatr 2010; 22: 405–411.
13. Edge J, Acerini C, Campbell F, et al. An alternative sensor-based method for glucose monitoring in children and young people with diabetes. Arch Dis Child 2017; 102: 543–549.
14. Buckingham BA, Raghinaru D, Cameron F, et al.; In Home Closed Loop Study Group. Predictive low-glucose insulin suspension reduces duration of nocturnal hypoglycemia in children without increasing ketosis. Diabetes Care 2015; 38: 1197–204.
15. Bergenstal RM, Garg S, Weinzimer SA, et al. Safety of a hybrid closed-loop insulin delivery system in patients with type 1 diabetes. JAMA 2016 4; 316: 1407–1408.
16. Riddell MC, Gallen IW, Smart CE, et al. Exercise management in type 1 diabetes: a consensus statement. Lancet Diabetes Endocrinol 2017; 5: 377–390.
17. Adolfsson P, Riddell MC, Taplin CE, et al. ISPAD Clinical Practice Consensus Guidelines 2018: exercise in children and adolescents with diabetes. Pediatr Diabetes 2018; 19 (Suppl 27): 205–226.
18. Messer LH, Forlenza GP, Wadwa RP, et al. The dawn of automated insulin delivery: A new clinical framework to conceptualize insulin administration. Pediatr Diabetes 2018; 19: 14–17.
19. Sherr JL, Tauschman M, Battelino T, et al. ISPAD clinical practice consensus guidelines 2018 Diabetes Technologies. Pediatr Diabetes, 2018; 19 Suppl 27: 302–325.
20. Bohn B, Herbst A, Pfeifer M, et al.; DPV Initiative. Impact of physical activity on glycemic control and prevalence of cardiovascular risk factors in adults with type 1 diabetes: a cross-sectional multicenter study of 18,028 patients. Diabetes Care 2015; 38: 1536–1543.
21. Metcalf KM, Singhvi A, Tsalikian E, et al. Effects of moderate-to-vigorous intensity physical activity on overnight and next-day hypoglycemia in active adolescents with type 1 diabetes. Diabetes Care 2014; 37: 1272–1278.
22. Smalley E. Medtronic automated insulin delivery device gets FDA nod. Nat Biotechnol 2016; 34: 1220.
23. de Boer IH, Gao X, Cleary PA, et al. Albuminuria changes and cardiovascular and renal outcomes in type 1 diabetes: the DCCT/EDIC Study. Clin J Am Soc Nephrol 2016; 11: 1969–1977.
24. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HAW. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359: 1577–1589.
25. Zoungas S, Chalmers J, Neal B, et al. Follow-up of blood-pressure lowering and glucose control in type 2 diabetes. N Engl J Med 2014; 371: 1392–406.
26. Mathieu C, Zinman B, Hemmingsson JU, et al.; ADJUNCT ONE Investigators. Efficacy and safety of liraglutide added to insulin treatment in type 1 diabetes: the ADJUNCT ONE treat-to-target randomized trial. Diabetes Care 2016; 39: 1702–1710.
27. Kuhadiya ND, Ghanim H, Mehta A, et al. Dapagliflozin as additional treatment to liraglutide and insulin in patients with type 1 diabetes. J Clin Endocrinol Metab 2016; 101: 3506–3515.
28. Inzucchi SE, Maggs DG, Spollett GR, et al. Efficacy and metabolic effects of metformin and troglitazone in type II diabetes mellitus. N Engl J Med 1998; 338: 867–872.
29. Abbasi F, Chu JW, McLaughlin T, Lamendola C, Leary ET, Reaven GM. Effect of metformin treatment on multiple cardiovascular disease risk factors in patients with type 2 diabetes mellitus. Metabolism 2004; 53: 159–164.
30. Libman IM, Miller KM, DiMeglio LA, et al.; T1D Exchange Clinic Network Metformin RCT Study Group. Effect of Metformin added to insulin on glycemic control among overweight/obese adolescents with type 1 diabetes: a randomized clinical trial. JAMA 2015; 1; 314: 2241–2250.
31. Al Khalifah RA, Alnhdi A, Alghar H, Alanazi M, Florez ID. The effect of adding metformin to insulin therapy for type 1 diabetes mellitus children: a systematic review and meta-analysis. Pediatr Diabetes 2017; 18: 664–673.
32. Shah AS, Dabelea D, Fino NF, et al. Predictors of increased carotid intima-media thickness in youthwith type 1 diabetes: the SEARCH CVD study. Diabetes Care 2016; 39: 418–425.
33. Nathan DM, Lachin J, Cleary P, et al.; Diabetes Control and Complications Trial Epidemiology of Diabetes Interventions and Complications Research Group. Intensive diabetes therapy and carotid intima media thickness in type 1 diabetes mellitus. N Engl J Med 2003; 348: 2294–2303.
34. Petitti DB, Klingensmith GJ, Bell RA, et al. Glycemic control in youth with diabetes: the SEARCH for Diabetes in Youth study. J Pediatr 2009; 155: 668–672.
35. Nambam B, Silverstein J, Cheng P, et al. A cross-sectional view of the current state of treatment of youth with type 2 diabetes in the enrollment data from the Pediatric Diabetes Consortium Type 2 Diabetes Registry. Pediatr Diabetes 2017; 18: 222–229.
36. Insel RA, Dunne JL, Atkinson MA, et al. Staging presymptomatic type 1 diabetes: a scientific statement of JDRF, the Endocrine Society, and the American Diabetes Association. Diabetes Care 2015; 38: 1964–1974.
Information & Authors
Information
Published In
Diabetes Technology & Therapeutics
Volume 21 • Issue Number S1 • February 2019
Pages: S-123 - S-137
PubMed: 30785328
Copyright
Copyright 2019, Mary Ann Liebert, Inc., publishers.
History
Published online: 20 February 2019
Published in print: February 2019
Topics
Authors
Author Disclosure Statement
D.M.M. has research support from the NIH, JDRF, NSF, and the Helmsley Charitable Trust, and his institution has research support from Medtronic, Dexcom, Insulet, Bigfoot Biomedical, Tandem, and Roche. He has consulted for Abbott, the Helmsley Charitable Trust, Sanofi, Novo Nordisk, Eli Lilly, and Insulet.
R.L. is a Stephen Bechtel Endowed Adult and Pediatric Endocrinology Fellow through the Stanford Child Health Research Institute and is supported by a Diabetes, Endocrinology and Metabolism Training Grant (T32 DK007217) from the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK). Dr. Lal has consulted for GlySens Incorporated and Abbott Diabetes Care.
S.S. has no competing financial interests.
Metrics & Citations
Metrics
Citations
Export Citation
Export citation
Select the format you want to export the citations of this publication.
View Options
View options
PDF/EPUB
View PDF/ePubGet Access
Access content
To read the fulltext, please use one of the options below to sign in or purchase access.⚠ Society Access
If you are a member of a society that has access to this content please log in via your society website and then return to this publication.