Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Nov 2.
Published in final edited form as: J Pediatr. 2015 Nov 5;169:13–20. doi: 10.1016/j.jpeds.2015.10.015

Progress in Diabetes Technology: Developments in Insulin Pumps, Continuous Glucose Monitors, and Progress towards the Artificial Pancreas

Gregory P Forlenza 1, Bruce Buckingham 2, David M Maahs 1
PMCID: PMC6214345  NIHMSID: NIHMS990937  PMID: 26547403

Type 1 diabetes (T1D) is one of the most common chronic diseases of childhood with an estimated prevalence of more than 166 000 cases in children younger than 20 years of age in the US in 2010.1 The rate of new-onset diabetes also is increasing worldwide, with an increased incidence of 3%-5% per year.2 In 1993 the Diabetes Control and Complications Trial established the benefit of intensive insulin therapy in reducing long-term complications, including retinopathy, nephropathy, and neuropathy.3 Despite this now longstanding knowledge, vascular complications attributable to hyperglycemia remain a significant issue in the population with T1D, even in the young-adult population.4,5 Recent evidence from the T1D Exchange clinic registry shows that during childhood mean hemoglobin A1c (HbA1c) remains above the target of 7.5% for all age groups, with a peak of 9.2% in the late teenage years.6 This evidence points toward the urgent need for better therapeutic interventions to improve glycemic control across the pediatric population. Innovations in diabetes care are being pursued on many fronts, including education, behavioral interventions, pharmaceutical development, beta-cell transplantation, and immunomodulation to prevent autoimmune beta-cell destruction. Although these therapies aim to provide benefit now and more so in the future, many in the diabetes community believe that the most impactful near-term benefit will be achieved by innovation in the technologies used to manage diabetes.

Portable subcutaneous continuous insulin infusion (CSII) pumps first became possible in the early 1970s and achieved improved glycemic control in early studies.7 Publication of the Diabetes Control and Complications Trial in 1993 demonstrated the importance of strict glycemic control and with it increased interest in the use of technology to minimize hyperglycemia without increasing hypoglycemia, still considered the greatest barrier to tight glucose control.3,8 The late 1990s and early 2000s saw rapid expansion in design and availability of CSII systems as research showed improved outcomes with use of this technology.9

The concept of a mechanical artificial pancreas has evolved with development of CSII pump and continuous glucose monitoring (CGM) technology. Such a system involves multiple components, including a continuous insulin delivery device, glucose sensor, insulin dosing decision algorithm, and components necessary for device communication. There has been some interest in development of devices that can sample intravenous blood and/or deliver intravenous or intraperitoneal insulin, although the major focus of research and development has been on subcutaneous glucose monitoring and subcutaneous insulin delivery systems. This interest fueled development of CGM systems during the 1990s, with the first commercial CGM device being approved in 1999.1015 Continued improvements in CGM technology facilitated both direct benefits to the care of patients with T1D and paved the way toward the development of emerging artificial pancreas systems. In this medical progress report, we review recent advances in diabetes technology, including CSII pumps, CGM systems, and emerging artificial pancreas technology.

CSII Pumps

During the past 20 years, CSII pump therapy has evolved as a mainstay for many patients with T1D. Retrospective crossover studies from the mid-2000s showed that switching from multiple daily injection (MDI) to CSII pump therapy was associated with significant improvement in average HbA1c (0.25%-0.75% reduction after 1 year of pump therapy), fasting blood glucose, episodes of hypoglycemia, and blood glucose variability, without increased episodes of diabetic ketoacidosis (DKA).1619 Of note, the greatest benefits were shown for the patients with the greatest baseline HbA1c before initiation of pump therapy, suggesting that the idea of patients showing good control to “earn a pump” may not be an optimal strategy. It is also worth noting that not all studies on this subject have demonstrated improved diabetes control with CSII therapy, and randomized prospective studies have not shown a benefit in younger children.2022 No increases in DKA with pump use compared with injections are seen in registry data from 5 different countries in>54 000 youth with T1D23 and children who have recurrent admissions to the hospital have a significant decrease in admissions when CSII therapy is initiated24

Studies on the durability of insulin pump use have continued to demonstrate superior glycemic control, lower insulin requirements, better health-related quality of life, and decreased hypoglycemic risk.25,26 Among adolescents, the greatest benefits are seen for those patients who use the advanced features of the devices.26

Additional emerging research has investigated psychosocial factors involved with insulin pump therapy. Patients generally identify a desire for improved glycemic control and flexibility of insulin dosing as reasons for transitioning from MDI to CSII pump therapy.27 Patient factors identified as predicting a greater rate of technology usage include a more active approach to diabetes care, realistic expectations of pump use, and recall of negative feelings at diabetes diagnosis.28 Factors identified as predicting a lower rate of use include a passive approach to self-care, with a view of the pump as an automatic-cure all.28 Detrimental aspects to pump use include body-image issues related to pump visibility, possible activity restrictions with pump use (eg, swimming with the pump), and concern over pump-site dysfunction and resultant DKA.27,28

Despite these noted benefits, <50% of patients in the US and Western Europe currently use insulin pumps. Data from 2011-2012 from 3 large clinical registries from the US and Western Europe shows that only 14% of patients in England and Wales, 41% of patients in Germany and Austria, and 47% of patients in the US were using insulin pumps.23 The use of a pump was associated with an HbA1c 0.5% lower than not using a pump.

A particular challenge among providers who care for patients with T1D is keeping up with the rapidly changing face of diabetes technology. Not only do products offered by established technology companies change every few years, but there is a frequent flux of companies entering and exiting the marketplace of diabetes technology. Most providers do not believe that it is their place to dictate to their patients which device they should purchase or to call one device “the best.” Rather, we should present the spectrum of available options with the relative strengths and weaknesses. Here we present a review of commercially available CSII devices for sale in the US (Table I). This list is by no means exhaustive but provides a general overview and comparison of commercially available products at the time of submission of this review.

Table I.

Commercially available insulin pumps, US, 2015

Pump brand CGM pairing CGM on-screen Low-glucose suspend Color screen Basal increments, U/hr Basal intervals, min Minimum bolus increments, U Tubing connection Blood glucose meter pairing
Animas Vibe (Animas Diabetes, West Chester, Pennsylvania) Yes; Dexcom G4 Yes No Yes 0.025 30 0.05 Luerlock None
Medtronic MiniMed Paradigm Revel 530G (Medtronic Diabetes, Northridge, California) Yes; Enlite 2 Yes Yes; Threshold Suspend No 0.025 30 0.025 Medtronic Bayer Contor Next
Insulet OmniPod (Insulet, Billerica, Massachusetts) Under development; Dexcom N/A No Yes 0.05 30 0.05 N/A - Tubeless Freestyle Integrated into PDM
Roche Accu-Chek Spirit Combo (Roche Diagnostics, Indianapolis, Indiana) Under development N/A No No 0.01 60 0.1 Luer lock Accu-Chek Aviva Combo
Tandem t:slim (Tandem Diabetes Care, San Diego, California) Yes; Dexcom G4 Yes No Yes; Touch 0.001 1 0.01 Luer lock None

N/A, not available; PDM, Personal Diabetes Manager.

CGM

CGM development represents a vital component in advancement in the clinical utility of diabetes technology. The presence of accurate real-time glucose values allows patients and their providers exponentially more data for diabetes care decisions. CGM data provides 24-hour tracking of blood glucose values as opposed to a focused snap-shot data provided by intermittent blood glucose meter testing. The combination of patient use of CGM and CSII pump technology with the patient/parent making all decisions in insulin dosing is known as sensor-augmented pump (SAP) therapy.29

Early studies on the benefits of CGM use showed significant benefits for young adults but not for teens or younger children, mostly attributable to the low rates of use in the younger age groups.30 Factors associated with greater rates of CGM use were patient age and frequency of blood glucose monitoring before beginning CGM and that frequency of CGM use predicted improvement in HbA1c.31 Among those patients in the younger age groups who used CGM frequently (≥6 d/wk) benefits were similar to those in the older age groups.32 Over an additional year, CGM use decreased with time in all age-groups, although frequent use of CGM was still associated with better HbA1c and reduced hypoglycemia in this cohort.3234

Investigation into patients’ attitudes and beliefs surrounding CGM is an emerging area of research. These studies show positive themes of improved blood glucose control, reduced worry/uncertainty, and improved overnight control; negative themes include: uncertainty with interpreting CGM data, lack of device accuracy, intrusiveness of alarms, and discomfort with wearing the device.35,36

Despite the noted benefits, overall use of CGM in clinical practice still remains quite low. Recent T1D Exchange data shows increasing rates of CGM usage among participants in this cohort; as of 2015, 11% of participants had used CGM,6 which is actually a 2% increase per year from 9% in the 2013-2014 report34 and a 7% rate of use among children in the 2010 report,6 possibly indicating increased uptake over time with improving technology. Among those who had used CGM, 41% of participants discontinued its use within 1 year, although it should be noted that this study was performed on an older generation of CGMs.34

Emerging CGM technologies hold significant potential to reverse this trend. Early-generation CGM systems used devices with lower accuracy, larger size, fewer features, and more frequent calibration than the current models. There are currently only 2 CGM devices on the market in the US (Table II). An additional system, the Abbott FreeStyle Navigator, was sold previously in the US, and a new generation of this technology, the FreeStyle Libre, recently has been approved in Europe.37

Table II.

Commercially available CGMs, US, 2015

CGM brand Pump pairing Sensor life, d Angle of insertion, degrees Warm-up time, h Calibration Glucose display frequency, min Directional trend display Remote monitoring available
Dexcom G4 PLATINUM and G5 Mobile (Dexcom, San Diego, California) Own receiver; Animas Vibe and t:slim; under development-OmniPod 7 45 2 Every 12 h 5 Yes Dexcom Share
Medtronic Enlite (Medtronic Diabetes, Northridge, California) Medtronic Paradigm 6 90 2 Every 12 h 5 Yes CareLink Connect

An emerging feature of CGM technology is the availability of remote monitoring, which has been accelerated by parent/patient advocates such as the Nightscout movement.38 This feature uses WiFi or cell phone signals to upload CGM data to cloud storage, enabling parents/caregivers to monitor a child’s blood sugar while the child and parent are separated (eg, school and work). These products have now begun to move to commercial release, with the Dexcom Share approved in January 2015 and the Medtronic CareLink Connect with projected approval in late 2015. The technical limitations of the current CGM devices include the need for multiple daily calibrations, pressure induced attenuation of sensor signal,39 interference from medications including acetaminophen,40 and current 7-day approved life of the sensor. The next generation of CGM sensors are reported to be addressing all of these issues, most notably coming “out of the box” with accurate factory calibration as with current blood glucose meter strips, eliminating the need for finger-stick calibration. The Abbott FreeStyle Libre is approved for use in Europe without calibration.37 Currently, the Food and Drug Administration does not approve of insulin dosing based solely on CGM data.

An additional limitation to more widespread CGM use is limited insurance coverage.41 However, a recent economic analysis of SAP use in Sweden showed a favorable cost-effectiveness ratio of SAP, mostly attributable to reduced incidence of diabetes-related complications.42 An older study from the Juvenile Diabetes Research Foundation (JDRF) also found long-term favorability of CGM use with an assumption of 2 fingerstick blood glucose tests per day.43 A more recent analysis of cost-effectiveness analysis outlines the benefits of CGM beyond HbA1c to include lowering hypoglycemia rates and the costs associated with them, both psychological and financial.41 Calibration-free devices could shift this equation towards the favorability of CGM as a cost-effective care tool as the cost of the sensor would be offset by decreased use of blood glucose meter test-strips. This shift in the cost-benefit equation would be most dramatically changed by approval for direct insulin dosing from CGM rather than from meter blood glucose values.

Artificial Pancreas Development

In 2006, the JDRF Artificial Pancreas project began, and in 2009 the JDRF outlined a 6 step-wise roadmap to development, refinement, and regulatory approval of a subcutaneous glucose monitoring and subcutaneous insulin delivery artificial pancreas system.44,45 This roadmap described successive steps from SAP therapy to systems involving sensor-directed suspension of insulin delivery to systems of hybrid and full closed-loop therapy and finally multihormone (eg, insulin and glucagon) therapy. Data exist on all steps of the 2009 roadmap summarized in this section, which demonstrate the feasibility of each.4655 Therefore, an updated roadmap has been introduced by Kowalski55 to emphasize a bifurcated pathway to insulin only and bihormonal insulin systems. The Food and Drug Administration also has issued updated guidelines specifically for artificial pancreas development with an attempt to achieve “the least burdensome approach” toward artificial pancreas testing and approval.56 They recommend that artificial pancreas systems be studied in 2 phases: first feasibility and then pivotal studies. Feasibility studies are underway and being completed at the time of this writing and pivotal studies are under design and implementation.

At the present time a step-1 commercial device (the Medtronic 530G system) with Threshold Suspend is available in the US and is helpful for patients with overnight hypoglycemia. This system successfully reduced overnight hypoglycemic events without producing rebound hyperglycemia in a recent phase 4 study.57 A step-2 commercial device (the Medtronic 640G system) with predictive low-glucose suspend was approved in Australia and in Europe. Initial studies that use a different predictive low glucose suspend algorithm have shown substantial reduction in overnight hypoglycemia in patients as young as 4 years of age.48,49,58 Step-3 devices with combined hypoglycemia and hyperglycemia minimizers are currently under phase 3 study.59

Devices consisting of hybrid closed-loop systems, fully automated closed-loop systems, and dual-hormonal systems are all under development at various stages of clinical testing at roughly a dozen centers around the world. The pathway to commercial approval for these advanced devices generally starts with in silico testing with computer-based compartment models of glucose response to insulin, followed by testing in a hospital setting, then in a controlled environment outside the hospital (eg, hotels and diabetes camps), and finally testing in the home environments.

Because of the pharmacokinetic limitations of current rapid acting insulins, closed-loop systems generally perform better during periods of fasting (such as overnight) and still have difficulty with the glycemic excursion of unannounced meals. Hybrid systems involve manual bolusing for meals and also use algorithm determined insulin delivery based on sensor glucose levels. These systems are especially effective overnight.52,60,61 A full closed-loop system would not require the user to enter meal boluses and would deliver all insulin without the need for the patient to enter food or exercise events.

A fundamental aspect of any artificial pancreas system is the control algorithm responsible for making dynamic insulin dosing adjustments in real-time. The theory for artificial pancreas algorithms arises from the discipline of control theory and dynamical systems.62,63 Control theory arises from ideas in mathematics and is a fundamental tool in many engineering disciplines, including aerospace, mechanical, chemical, and electrical and computer engineering. A comprehensive review of the engineering of artificial pancreas algorithms was recently published by Doyle et al.64

Groups around the world generally are using 3 different control systems for Closed Loop (CL) therapy: proportional-integral-derivative (PID) control, model predictive control (MPC), and fuzzy logic control. PID control is probably the most basic form of a control system. At each point in time the controller assesses how far the current glucose is from the desired glucose (proportional), the rate of change in glucose (derivative), and how long the glucose has remained above or below target (integral), and then uses a weighted sum of these factors to determine an insulin dose for that point in time.52,65 MPC relies on the development of a complex multicompartment model via a series of differential equations. This model is then used to predict the appropriate dosing action for a fixed time interval (eg, 5-15 minutes), after which time the system is reassessed and the appropriate model selected for the next time interval.66,67 Fuzzy logic control uses a series of “fuzzy” logical decision rules based on the current glucose and the direction and rate of change of the glucose to make reasoned decisions on insulin doses.68,69 A fuzzy logic controller attempts to imitate the reasoning of a diabetes clinician whereby an expert clinician’s dosing expertise is codified in terms of dosing rules based on different glucose circumstances such as glucose level, glucose rate of change, and glucose acceleration.70 In addition to commercial development of artificial pancreas systems, patient/parent-driven development is also occurring in a manner similar to the Nightscout movement (http://www.nightscoutfoundation.org/about) with CGM. Bigfoot Biomedical is a startup company with a CL algorithm developed by parents of children with T1D that intends to drive artificial pancreas development rapidly forward.71 Do It Yourself Pancreas System started as a couple hacking the CGM to make the alarms louder and has now progressed into a patient-driven artificial pancreas system that uses Open Source artificial pancreas code.72

Current research on CL systems demonstrate an overall common theme of successful control of overnight blood glucose with values in target range 70%-100% of the time, but there is some difficulty in preventing postmeal glycemic excursions with unannounced meals. A summary article of CL research was published recently by Shah et al.73 Research on MPC systems has shown increased time in normal range without increased hyperglycemia in hospital settings when compared with MDI and SAP therapy,7479 and has now shown reduced hypoglycemia and increased time in range in early camp and out-patient settings.50,61,7981 PID systems have shown improved time in range with reduced hypoglycemia in hospital and camp settings.60,82 Fuzzy logic systems have shown increased time in range with reduced hypoglycemia compared with SAP in outpatient settings.83 The “Bionic Pancreas” provides the replacement of both insulin and glucagon.84,85 This system has been used successfully in a hotel setting and in diabetes camps, achieving an average glucose in adults/adolescents of 138/138 mg/dL with 4.1/6.1% of readings <70 mg/dL. Studies combining closed-loop artificial pancreas technology with adjuvant diabetes medication also are underway. These studies investigate the role of adjuvant medications such as sodium-glucose linked transporter 2 inhibitors, combined sodium-glucose linked transporter 1/2 inhibitors, amylin analogues, glucagon-like peptide-1 agonists, and inhaled insulin. Studies combining these agents with diabetes technology show early promise to help mitigate postprandial hyperglycemia.8690

Barriers on the Pathway to Clinical Use

There are several commonly cited technical barriers to full implementation of CL technology. These include: (1) the need for more accurate CGM devices with less calibration, better accuracy, and longer life-span; (2) the need for “ultra-rapid” insulins that have a faster onset, more rapid peak, and shorter duration of action to more closely replicate pancreatic portal insulin onset and duration of action particularly as it pertains to preventing meal-time glycemic excursions and postprandial hyperglycemia; (3) the continued need for algorithm refinement and improvement possibly including features for exercise, stress, sleep, and illness; and (4) the need for continued improvement in the user interface which includes: (a) the device interfaces with the user; (b) device connectivity between sensors, pumps, the controller, and the cloud; and (c) the sites at which devices are worn on the skin, which includes tape, insertion sites, the number of devices worn, and the length of time that they can be worn. An additional technical barrier for dual-hormone (insulin and glucagon) systems is the lack of a commercially available aqueous stable formulation of glucagon. Currently commercially available glucagon forms fibrils in aqueous solution and is therefore only usable for 24 hours after reconstitution. In response to this limitation commercial development is underway on stable aqueous and nonaqueous glucagon formulations.91,92

One of the major focuses on future technological development must be the need to reduce the burden of T1D. The concept of reduced burden has begun to emerge in the technology community and its success will likely be linked with the future uptake and success of artificial pancreas technology.93 In his recent pathway update, Kowalski55 addresses these barriers as well as the needs for patient and health care provider acceptance and payer coverage of emerging diabetes technology. The concept of cost-effectiveness of advanced diabetes technologies has also begun to be studied. Although upfront costs for these systems will be significant, predictive economic analysis has shown that reduced hospital costs from visits to the emergency department and admissions caused by hypoglycemia and long-term complication reduction from improved HbA1c show a cost benefit for advanced diabetes technology including artificial pancreas systems.41,94 Cost-effectiveness of these systems is critical as, the ultimate metric for success of artificial pancreas systems will be based on their widespread use and how they improve quality of life, reduce stress, and improve outcomes for all patients with diabetes.

Conclusions

All steps in the artificial pancreas pathway have been shown to be feasible. Studies are now moving from preliminary controlled in-patient settings to pivotal real-world outpatient settings. Continued development of these technologies must focus on patient-centered needs and reducing the global burden of T1D on these patients. With this focus, diabetes technology promises to reduce burden and improve clinical outcomes for a wide spectrum of patients with diabetes in the near future.

Acknowledgments

G.F. receives research supplies from Medtronic. B.B. serves as a Medical Advisory Board consultant for Medtronic, Sanofi, Tandem, and Novo-Nordisk; and receives financial support from Medtronic, Dexcom, and BD. D.M. receives research support from Medtronic and Dexcom; and serves on the scientific advisory board for Insulet.

Glossary

CGM

Continuous glucose monitoring

CSII

Continuous insulin infusion

DKA

Diabetic ketoacidosis

HbA1c

Hemoglobin A1c

JDRF

Juvenile Diabetes Research Foundation

MDI

Multiple daily injection

MPC

Multiple daily injection

PID

Proportional-integral-derivative

SAP

Sensor-augmented pump

T1D

Type 1 diabetes

References

  • 1.Pettitt DJ, Talton J, Dabelea D, Divers J, Imperatore G, Lawrence JM, et al. Prevalence of diabetes in U.S. youth in 2009: the SEARCH for diabetes in youth study. Diabetes Care 2014;37:402–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Forlenza GP, Rewers M. The epidemic of type 1 diabetes: what is it telling us? Curr Opin Endocrinol Diabetes Obes 2011;18:248–51. [DOI] [PubMed] [Google Scholar]
  • 3.The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med 1993;329:977–86. [DOI] [PubMed] [Google Scholar]
  • 4.James S, Gallagher R, Dunbabin J, Perry L. Prevalence of vascular complications and factors predictive of their development in young adults with type 1 diabetes: systematic literature review. BMC Res Notes 2014;7:593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Maahs DM, Daniels SR, de Ferranti SD, Dichek HL, Flynn J, Goldstein BI, et al. Cardiovascular disease risk factors in youth with diabetes mellitus: a scientific statement from the american heart association. Circulation 2014;130:1532–58. [DOI] [PubMed] [Google Scholar]
  • 6.Miller KM, Foster NC, Beck RW, Bergenstal RM, DuBose SN, DiMeglio LA, et al. Current state of type 1 diabetes treatment in the U.S.: updated data from the T1D Exchange Clinic Registry. Diabetes Care 2015;38:971–8. [DOI] [PubMed] [Google Scholar]
  • 7.Tamborlane WV, Sherwin RS, Genel M, Felig P. Reduction to normal of plasma glucose in juvenile diabetes by subcutaneous administration of insulin with a portable infusion pump. N Engl J Med 1979;300:573–8. [DOI] [PubMed] [Google Scholar]
  • 8.Cryer PE. Mechanisms of hypoglycemia-associated autonomic failure in diabetes. N Engl J Med 2013;369:362–72. [DOI] [PubMed] [Google Scholar]
  • 9.Kaufman FR, Halvorson M, Miller D, Mackenzie M, Fisher LK, Pitukcheewanont P. Insulin pump therapy in type 1 pediatric patients: now and into the year 2000. Diabetes Metab Res Rev 1999;15:338–52. [DOI] [PubMed] [Google Scholar]
  • 10.Shichiri M, Yamasaki Y, Nao K, Sekiya M, Ueda N. In vivo characteristics of needle-type glucose sensor–measurements of subcutaneous glucose concentrations in human volunteers. Horm Metab Res Suppl 1988;20: 17–20. [PubMed] [Google Scholar]
  • 11.Pfeiffer EF. The glucose sensor: the missing link in diabetes therapy. Horm Metab Res Suppl 1990;24:154–64. [PubMed] [Google Scholar]
  • 12.Fischer U Continuous in vivo monitoring in diabetes: the subcutaneous glucose concentration. Acta Anaesthesiol Scand Suppl 1995;104:21–9. [DOI] [PubMed] [Google Scholar]
  • 13.Ginsbergh BH. The FDA panel advised approval of the first continuous glucose sensor. Diabetes Technol Ther 1999;1:203–4. [DOI] [PubMed] [Google Scholar]
  • 14.Rebrin K, Steil GM, van Antwerp WP, Mastrototaro JJ. Subcutaneous glucose predicts plasma glucose independent of insulin: implications for continuous monitoring. Am J Physiol 1999;277:E561–71. [DOI] [PubMed] [Google Scholar]
  • 15.Rebrin K, Fischer U, Hahn von Dorsche H, Von Woetke T, Abel P, Brunstein E. Subcutaneous glucose monitoring by means of electrochemical sensors: fiction or reality. J Biomed Eng 1992;14:33–40. [DOI] [PubMed] [Google Scholar]
  • 16.Berhe T, Postellon D, Wilson B, Stone R. Feasibility and safety of insulin pump therapy in children aged 2 to 7 years with type 1 diabetes: a retrospective study. Pediatrics 2006;117:2132–7. [DOI] [PubMed] [Google Scholar]
  • 17.Nimri R, Weintrob N, Benzaquen H, Ofan R, Fayman G, Phillip M. Insulin pump therapy in youth with type 1 diabetes: a retrospective paired study. Pediatrics 2006;117:2126–31. [DOI] [PubMed] [Google Scholar]
  • 18.Retnakaran R, Hochman J, DeVries JH, Hanaire-Broutin H, Heine RJ, Melki V, et al. Continuous subcutaneous insulin infusion versus multiple daily injections: the impact of baseline A1c. Diabetes Care 2004;27:2590–6. [DOI] [PubMed] [Google Scholar]
  • 19.Weissberg-Benchell J, Antisdel-Lomaglio J, Seshadri R. Insulin pump therapy: a meta-analysis. Diabetes Care 2003;26:1079–87. [DOI] [PubMed] [Google Scholar]
  • 20.DiMeglio LA, Pottorff TM, Boyd SR, France L, Fineberg N, Eugster EA. A randomized, controlled study of insulin pump therapy in diabetic preschoolers. J Pediatr 2004;145:380–4. [DOI] [PubMed] [Google Scholar]
  • 21.Fox LA, Buckloh LM, Smith SD, Wysocki T, Mauras N. A randomized controlled trial of insulin pump therapy in young children with type 1 diabetes. Diabetes Care 2005;28:1277–81. [DOI] [PubMed] [Google Scholar]
  • 22.Nabhan ZM, Kreher NC, Greene DM, Eugster EA, Kronenberger W, DiMeglio LA. A randomized prospective study of insulin pump vs. insulin injection therapy in very young children with type 1 diabetes: 12-month glycemic, BMI, and neurocognitive outcomes. Pediatr Diabetes 2009;10:202–8. [DOI] [PubMed] [Google Scholar]
  • 23.Maahs DM, Hofer SE, Foster NC, Hermann JM, Tamborlane WV, Kapellen TM, et al. Insulin pump use in pediatric type 1 diabetes: multinational comparison with 54,768 pediatric patients from the T1D exchange (US), national paediatric diabetes audit (England and Wales), and the DPV initiative (Germany and Austria) [abstract]. Pediatr Diabetes 2014;15:47.25182307 [Google Scholar]
  • 24.Steindel BS, Roe TR, Costin G, Carlson M, Kaufman FR. Continuous subcutaneous insulin infusion (CSII) in children and adolescents with chronic poorly controlled type 1 diabetes mellitus. Diabetes Res Clin Pract 1995;27:199–204. [DOI] [PubMed] [Google Scholar]
  • 25.Pozzilli P, Battelino T, Danne T, Hovorka R, Jarosz-Chobot P, Renard E. Continuous subcutaneous insulin infusion in diabetes: patient populations, safety, efficacy, and pharmacoeconomics. Diabetes Metab Res Rev 2015. April 13 10.1002/dmrr.2653 [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mameli C, Scaramuzza AE, Ho J, Cardona-Hernandez R, Suarez-Ortega L, Zuccotti GV. A 7-year follow-up retrospective, international, multicenter study of insulin pump therapy in children and adolescents with type 1 diabetes. Acta Diabetol 2014;51:205–10. [DOI] [PubMed] [Google Scholar]
  • 27.Alsaleh FM, Smith FJ, Taylor KM. Experiences of children/young people and their parents, using insulin pump therapy for the management of type 1 diabetes: qualitative review. J Clin Pharm Ther 2012;37:140–7. [DOI] [PubMed] [Google Scholar]
  • 28.Ritholz MD, Smaldone A, Lee J, Castillo A, Wolpert H, Weinger K. Perceptions of psychosocial factors and the insulin pump. Diabetes Care 2007;30:549–54. [DOI] [PubMed] [Google Scholar]
  • 29.Bergenstal RM, Tamborlane WV, Ahmann A, Buse JB, Dailey G, Davis SN, et al. Effectiveness of sensor-augmented insulin-pump therapy in type 1 diabetes. N Engl J Med 2010;363:311–20. [DOI] [PubMed] [Google Scholar]
  • 30.Juvenile Diabetes Research Foundation Continuous Glucose Monitoring Study GroupTamborlane WV, Beck RW, Bode BW, Buckingham B, Chase HP, et al. Continuous glucose monitoring and intensive treatment of type 1 diabetes. N Engl J Med 2008;359:1464–76. [DOI] [PubMed] [Google Scholar]
  • 31.Beck RW, Buckingham B, Miller K, Wolpert H, Xing D, Block JM, et al. Factors predictive of use and of benefit from continuous glucose monitoring in type 1 diabetes. Diabetes Care 2009;32:1947–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Effectiveness of continuous glucose monitoring in a clinical care environment: evidence from the Juvenile Diabetes Research Foundation continuous glucose monitoring (JDRF-CGM) trial. Diabetes Care 2010;33:17–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chase HP, Beck RW, Xing D, Tamborlane WV, Coffey J, Fox LA, et al. Continuous glucose monitoring in youth with type 1 diabetes: 12-month follow-up of the Juvenile Diabetes Research Foundation continuous glucose monitoring randomized trial. Diabetes Technol Ther 2010; 12:507–15. [DOI] [PubMed] [Google Scholar]
  • 34.Wong JC, Foster NC, Maahs DM, Raghinaru D, Bergenstal RM, Ahmann AJ, et al. Real-time continuous glucose monitoring among participants in the T1D Exchange clinic registry. Diabetes Care 2014;37: 2702–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Polonsky WH, Hessler D. What are the quality of life-related benefits and losses associated with real-time continuous glucose monitoring? A survey of current users. Diabetes Technol Ther 2013;15:295–301. [DOI] [PubMed] [Google Scholar]
  • 36.Barnard KD, Wysocki T, Thabit H, Evans ML, Amiel S, Heller S, et al. Psychosocial aspects of closed- and open-loop insulin delivery: closing the loop in adults with Type 1 diabetes in the home setting. Diabet Med 2015;32:601–8. [DOI] [PubMed] [Google Scholar]
  • 37.Abbott. Newsroom: Abbott Receives CE Mark for FreeStyle_ Libre, a Revolutionary Glucose Monitoring System for People with Diabetes. https://www.abbottdiabetescare.com/press-room/2014.html Accessed October 15, 2015.
  • 38.Sparling K CGM in the Cloud: the how, why, and why not of remote CGM watching. http://diatribe.org/companiesorganization/nightscout#sthash.m9rNhBia.dpuf Accessed October 15, 2015. [Google Scholar]
  • 39.Baysal N, Cameron F, Buckingham BA, Wilson DM, Chase HP, Maahs DM, et al. A novel method to detect pressure-induced sensor attenuations (PISA) in an artificial pancreas. J Diabetes Sci Technol 2014; 8:1091–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Maahs DM, DeSalvo D, Pyle L, Ly T, Messer L, Clinton P, et al. Effect of acetaminophen on CGM glucose in an outpatient setting. Diabetes Care 2015;38:e158–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Vigersky RA. The benefits, limitations, and cost-effectiveness of advanced technologies in the management of patients with diabetes mellitus. J Diabetes Sci Technol 2015;9:320–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Roze S, Saunders R, Brandt A, de Portu S, Papo NL, Jendle J. Health-economic analysis of real-time continuous glucose monitoring in people with Type 1 diabetes. Diabet Med 2015;32:618–26. [DOI] [PubMed] [Google Scholar]
  • 43.Huang ES, O’Grady M, Basu A, Winn A, John P, Lee J, et al. The cost-effectiveness of continuous glucose monitoring in type 1 diabetes. Diabetes Care 2010;33:1269–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Juvenile Diabetes Research Foundation. Artificial Pancreas Project Research; 2006. http://jdrf.org/research/treat/artificial-pancreas-project/ Accessed March 25, 2015.
  • 45.Kowalski AJ. Can we really close the loop and how soon? Accelerating the availability of an artificial pancreas: a roadmap to better diabetes outcomes. Diabetes Technol Ther 2009;11(Suppl 1):S113–9. [DOI] [PubMed] [Google Scholar]
  • 46.Ly TT, Nicholas JA, Retterath A, Lim EM, Davis EA, Jones TW. Effect of sensor-augmented insulin pump therapy and automated insulin suspension vs standard insulin pump therapy on hypoglycemia in patients with type 1 diabetes: a randomized clinical trial. JAMA 2013;310:1240–7. [DOI] [PubMed] [Google Scholar]
  • 47.Bergenstal RM, Klonoff DC, Garg SK, Bode BW, Meredith M, Slover RH, et al. Threshold-based insulin-pump interruption for reduction of hypoglycemia. N Engl J Med 2013;369:224–32. [DOI] [PubMed] [Google Scholar]
  • 48.Maahs DM, Calhoun P, Buckingham BA, Chase HP, Hramiak I, Lum J, et al. A randomized trial of a home system to reduce nocturnal hypoglycemia in type 1 diabetes. Diabetes Care 2014;37:1885–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Buckingham BA, Cameron F, Calhoun P, Maahs DM, Wilson DM, Chase HP, et al. Outpatient safety assessment of an in-home predictive low-glucose suspend system with type 1 diabetes subjects at elevated risk of nocturnal hypoglycemia. Diabetes Technol Ther 2013;15:622–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kovatchev BP, Renard E, Cobelli C, Zisser HC, Keith-Hynes P, Anderson SM, et al. Safety of outpatient closed-loop control: first randomized crossover trials of a wearable artificial pancreas. Diabetes Care 2014;37:1789–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Leelarathna L, Dellweg S, Mader JK, Allen JM, Benesch C, Doll W, et al. Day and night home closed-loop insulin delivery in adults with type 1 diabetes: three-center randomized crossover study. Diabetes Care 2014;37:1931–7. [DOI] [PubMed] [Google Scholar]
  • 52.Weinzimer SA, Steil GM, Swan KL, Dziura J, Kurtz N, Tamborlane WV. Fully automated closed-loop insulin delivery versus semiautomated hybrid control in pediatric patients with type 1 diabetes using an artificial pancreas. Diabetes Care 2008;31:934–9. [DOI] [PubMed] [Google Scholar]
  • 53.Phillip M, Battelino T, Atlas E, Kordonouri O, Bratina N, Miller S, et al. Nocturnal glucose control with an artificial pancreas at a diabetes camp. N Engl J Med 2013;368:824–33. [DOI] [PubMed] [Google Scholar]
  • 54.Russell SJ, El-Khatib FH, Sinha M, Magyar KL, McKeon K, Goergen LG, et al. Outpatient glycemic control with a bionic pancreas in type 1 diabetes. N Engl J Med 2014;371:313–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kowalski A Pathway to artificial pancreas systems revisited: moving downstream. Diabetes Care 2015;38:1036–43. [DOI] [PubMed] [Google Scholar]
  • 56.Beck S Guidance for industry and food and drug administration staff: The content of investigational device exemption (IDE) and premarket approval (PMA) applications for artificial pancreas device systems [Internet] . Rockville (MD): U.S. Department of Health and Human Services; Food and Drug Administration; Center for Devices and Radiological Health; 2012. [Google Scholar]
  • 57.Agrawal P, Zhong A, Welsh JB, Shah R, Kaufman FR. Retrospective analysis of the real-world use of the threshold suspend feature of sensor-augmented insulin pumps. Diabetes Technol Ther 2015;17:316–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Buckingham BA, Raghinaru D, Cameron F, Bequette BW, Chase HP, Maahs DM, et al. Predictive low-glucose insulin suspension reduces duration of nocturnal hypoglycemia in children without increasing ketosis. Diabetes Care 2015;38:1197–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Finan DA, McCann TW Jr, Mackowiak L, Dassau E, Patek SD, Kovatchev BP, et al. Closed-Loop Control Performance of the Hypoglycemia-Hyperglycemia Minimizer (HHM) System in a Feasibility Study. J Diabetes Sci Technol 2014;8:35–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ly TT, Roy A, Grosman B, Shin J, Campbell A, Monirabbasi S, et al. Day and Night Closed-Loop Control Using the Integrated Medtronic Hybrid Closed-Loop System in Type 1 Diabetes at Diabetes Camp. Diabetes Care 2015;38:1205–11. [DOI] [PubMed] [Google Scholar]
  • 61.Ly TT, Breton MD, Keith-Hynes P, De Salvo D, Clinton P, Benassi K, et al. Overnight glucose control with an automated, unified safety system in children and adolescents with type 1 diabetes at diabetes camp. Diabetes Care 2014;37:2310–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Astrom KJ, Murray RM. Feedback systems: An introduction for scientists and engineers. Princeton, NJ: Princeton University Press; 2008. [Google Scholar]
  • 63.Bequette BW. Algorithms for a closed-loop artificial pancreas: the case for model predictive control. J Diabetes Sci Technol 2013;7:1632–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Doyle FJ III, Huyett LM, Lee JB, Zisser HC, Dassau E. Closed-loop artificial pancreas systems: engineering the algorithms. Diabetes Care 2014; 37:1191–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Steil GM, Rebrin K, Darwin C, Hariri F, Saad MF. Feasibility of automating insulin delivery for the treatment of type 1 diabetes. Diabetes 2006;55:3344–50. [DOI] [PubMed] [Google Scholar]
  • 66.Hovorka R, Canonico V, Chassin LJ, Haueter U, Massi-Benedetti M, Orsini Federici M, et al. Nonlinear model predictive control of glucose concentration in subjects with type 1 diabetes. Physiol Meas 2004;25:905–20. [DOI] [PubMed] [Google Scholar]
  • 67.Clarke WL, Anderson S, Breton M, Patek S, Kashmer L, Kovatchev B. Closed-loop artificial pancreas using subcutaneous glucose sensing and insulin delivery and a model predictive control algorithm: the Virginia experience. J Diabetes Sci Technol 2009;3:1031–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Nimri R, Phillip M. Artificial pancreas: fuzzy logic and control of glycemia. Curr Opin Endocrinol Diabetes Obes 2014;21:251–6. [DOI] [PubMed] [Google Scholar]
  • 69.Mauseth R, Wang Y, Dassau E, Kircher R Jr, Matheson D, Zisser H, et al. Proposed clinical application for tuning fuzzy logic controller of artificial pancreas utilizing a personalization factor. J Diabetes Sci Technol 2010;4: 913–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Mauseth R, Hirsch IB, Bollyky J, Kircher R, Matheson D, Sanda S, et al. Use of a “fuzzy logic” controller in a closed-loop artificial pancreas. Diabetes Technol Ther 2013;15:628–33. [DOI] [PubMed] [Google Scholar]
  • 71.diaTribe, Close K, Brown A. Bigfoot Biomedical Sets its Sights on Simplifying Type 1 Diabetes Management, http://diatribe.org/bigfoot-biomedical-sets-its-sights-simplifying-type-1-diabetes-management; 2015. Accessed October 15, 2015. [Google Scholar]
  • 72.Carson B. They hacked her pancreas and found love along the way, http://www.businessinsider.com/hacked-raspberry-pi-artificial-pancreas-2015-8?utm_source=gatehouse&utm_medium=referral&utm_content=feed;2015 Accessed October 15, 2015. [Google Scholar]
  • 73.Shah VN, Shoskes A, Tawfik B, Garg SK. Closed-loop system in the management of diabetes: past, present, and future. Diabetes Technol Ther 2014;16:477–90. [DOI] [PubMed] [Google Scholar]
  • 74.Breton M, Farret A, Bruttomesso D, Anderson S, Magni L, Patek S, et al. Fully integrated artificial pancreas in type 1 diabetes: modular closed-loop glucose control maintains near normoglycemia. Diabetes 2012;61: 2230–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Cameron F, Niemeyer G, Wilson DM, Bequette BW, Benassi KS, Clinton P, et al. Inpatient trial of an artificial pancreas based on multiple model probabilistic predictive control with repeated large unannounced meals. Diabetes Technol Ther 2014;16:728–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chase HP, Doyle FJ III, Zisser H, Renard E, Nimri R, Cobelli C, et al. Multicenter closed-loop/hybrid meal bolus insulin delivery with type 1 diabetes. Diabetes Technol Ther 2014;16:623–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Elleri D, Allen JM, Biagioni M, Kumareswaran K, Leelarathna L, Caldwell K, et al. Evaluation of a portable ambulatory prototype for automated overnight closed-loop insulin delivery in young people with type 1 diabetes. Pediatr Diabetes 2012;13:449–53. [DOI] [PubMed] [Google Scholar]
  • 78.Elleri D, Allen JM, Kumareswaran K, Leelarathna L, Nodale M, Caldwell K, et al. Closed-loop basal insulin delivery over 36 hours in adolescents with type 1 diabetes: randomized clinical trial. Diabetes Care 2013;36:838–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zisser H, Renard E, Kovatchev B, Cobelli C, Avogaro A, Nimri R, et al. Multicenter closed-loop insulin delivery study points to challenges for keeping blood glucose in a safe range by a control algorithm in adults and adolescents with type 1 diabetes from various sites. Diabetes Technol Ther 2014;16:613–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Brown SA, Kovatchev BP, Breton MD, Anderson SM, Keith-Hynes P, Patek SD, et al. Multinight “bedside” closed-loop control for patients with type 1 diabetes. Diabetes Technol Ther 2015;17:203–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Thabit H, Elleri D, Leelarathna L, Allen JM, Lubina-Solomon A, Stadler M, et al. Unsupervised home use of an overnight closed-loop system over 3-4 weeks: a pooled analysis of randomized controlled studies in adults and adolescents with type 1 diabetes. Diabetes Obes Metab 2015;17:452–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.O’Grady MJ, Retterath AJ, Keenan DB, Kurtz N, Cantwell M, Spital G, et al. The use of an automated, portable glucose control system for overnight glucose control in adolescents and young adults with type 1 diabetes. Diabetes Care 2012;35:2182–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Nimri R, Muller I, Atlas E, Miller S, Fogel A, Bratina N, et al. MDLogic overnight control for 6 weeks of home use in patients with type 1 diabetes: randomized crossover trial. Diabetes Care 2014;37: 3025–32. [DOI] [PubMed] [Google Scholar]
  • 84.El-Khatib FH, Jiang J, Damiano ER. Adaptive closed-loop control provides blood-glucose regulation using dual subcutaneous insulin and glucagon infusion in diabetic Swine. J Diabetes Sci Technol 2007;1: 181–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Jacobs PG, El Youssef J, Castle J, Bakhtiani P, Branigan D, Breen M, et al. Automated control of an adaptive bihormonal, dual-sensor artificial pancreas and evaluation during inpatient studies. IEEE Trans Biomed Eng 2014;61:2569–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Sands AT, Zambrowicz BP, Rosenstock J, Lapuerta P, Bode BW, Garg SK, et al. Sotagliflozin, a dual SGLT1 and SGLT2 inhibitor, as adjunct therapy to insulin in type 1 diabetes. Diabetes Care 2015;38: 1181–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Traina AN, Lull ME, Hui AC, Zahorian TM, Lyons-Patterson J. Once-weekly exenatide as adjunct treatment of type 1 diabetes mellitus in patients receiving continuous subcutaneous insulin infusion therapy. Can J Diabetes 2014;38:269–72. [DOI] [PubMed] [Google Scholar]
  • 88.Renukuntla VS, Ramchandani N, Trast J, Cantwell M, Heptulla RA. Role of glucagon-like peptide-1 analogue versus amylin as an adjuvant therapy in type 1 diabetes in a closed loop setting with ePID algorithm. J Diabetes Sci Technol 2014;8:1011–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Perkins BA, Cherney DZ, Partridge H, Soleymanlou N, Tschirhart H, Zinman B, et al. Sodium-glucose cotransporter 2 inhibition and glycemic control in type 1 diabetes: results of an 8-week open-label proof-of-concept trial. Diabetes Care 2014;37: 1480–3. [DOI] [PubMed] [Google Scholar]
  • 90.Zisser H, Dassau E, Lee JJ, Harvey RA, Bevier W, Doyle FJ III. Clinical results of an automated artificial pancreas using technosphere inhaled insulin to mimic first-phase insulin secretion. J Diabetes Sci Technol 2015;9:564–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Bakhtiani PA, Caputo N, Castle JR, El Youssef J, Carroll JM, David LL, et al. A novel, stable, aqueous glucagon formulation using ferulic acid as an excipient. J Diabetes Sci Technol 2015;9:17–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Newswanger B, Ammons S, Phadnis N, Ward WK, Castle J, Campbell RW, et al. Development of a highly stable, nonaqueous glucagon formulation for delivery via infusion pump systems. J Diabetes Sci Technol 2015;9:24–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Beck RW. Downloading diabetes device data: empowering patients to download at home to achieve better outcomes. Diabetes Technol Ther 2015;17:536–7. [DOI] [PubMed] [Google Scholar]
  • 94.O’Grady MJ, John P, Winn A. Substantial Medicare savings may result if insurers cover ‘artificial pancreas’ sooner for diabetes patients. Health Affairs (Project Hope) 2012;31:1822–9. [DOI] [PubMed] [Google Scholar]

RESOURCES