OBJECTIVE

Continuous glucose monitoring (CGM) combined with continuous subcutaneous insulin infusion (CSII) achieves better glycemic control than multi-injection therapy in people with type 2 diabetes. The effectiveness of closed-loop therapy needs to be further evaluated in this population.

RESEARCH DESIGN AND METHODS

The study objective was to measure the impact of a hybrid closed-loop device (DBLG1) compared with CSII + CGM on glycemic control in people with type 2 diabetes previously treated with CSII. The randomized, controlled, crossover, two-period, open-label, and multicenter study was conducted from August 2022 to July 2023 in 17 individuals (9 to receive 6 weeks of CSII + CGM first and 8 to receive 6 weeks of closed-loop therapy first). The primary end point was the percentage time in range (TIR: 70–180 mg/dL). Secondary outcomes were other CGM-glucose metrics, physical activity, and sleep objectively measured using 1-week actimetry.

RESULTS

Data were analyzed using a modified intention-to-treat approach. Mean age was 63 (SD 9) years and 35% were women. Mean HbA1c at inclusion was 7.9% (SD 0.9). TIR increased to 76.0% (interquartile range 69.0–84.0) during the closed-loop condition vs. 61.0% (interquartile range 55.0–70.0) during the CSII + CGM condition; mean difference was 15.0 percentage points (interquartile range 8.0–22.0; P < 0.001). Analyses of secondary end points showed a decrease in time above range, in glucose management indicator, in glucose variability, and an increase in daily insulin dose. Actimetric sleep analysis showed an improvement in sleep fragmentation during closed-loop treatment.

CONCLUSIONS

Closed-loop therapy improved glycemic control more than did CSII + CGM in people with type 2 diabetes.

The prevalence of type 2 diabetes has risen steadily since the 1980s. The International Diabetes Federation estimated that in 2021, 537 million people in the world had diabetes, and this number is projected to reach 643 million by 2030 and 783 million by 2045 (1). People who live with type 2 diabetes have a higher risk of organ dysfunction and failure, especially the kidneys, eyes, and nerves. It is estimated that people with type 2 diabetes have an excess mortality risk from any cause of 15% compared to general population and that this mortality risk increases substantially with poorer glycemic control, severe renal complications, impaired renal function, and younger age at diagnosis (2).

Type 2 diabetes is a progressive disease. Treatment needs to be intensified over time until insulin replacement becomes necessary. In France, the Echantillon National Témoin Représentatif des personnes Diabétiques, 3rd edition (ENTRED3) epidemiological study showed that 22.5% of people living with type 2 diabetes are treated with insulin (3). Several studies have shown improvements in glycemic control with continuous subcutaneous insulin infusion (CSII) when people with type 2 diabetes reach the stage of requiring multiple daily insulin injections (4,5). In addition, using continuous glucose monitoring (CGM) or flash CGM (FGM) in adults with type 2 diabetes has also improved glycemic control (6).

In people with type 1 diabetes, closed-loop therapy has been shown to be superior to glucose sensor-augmented pump therapy or any other treatment modality. Closed-loop systems allow more time to be spent in range (TIR, 70–180 mg/dL), reduce glycemic variability, and reduce the risk of hypoglycemia (7–11).

Insulin delivery by closed-loop therapy in type 2 diabetes has been tested in eight randomized controlled trials (RCTs) to date. Five of these were conducted in hospitalized individuals with treatment exposure times of <20 days (12–16). Three were conducted in outpatients with an exposure duration of 20 days (17), 8 weeks (18), and 12 weeks (19), respectively. Four trials were multicenter (13,14,17,19). One trial was conducted in people requiring nutritional support (14), another during the perioperative period of elective surgery (16), and another in people treated with dialysis (17). All of these trials (18) involved comparison with conventional insulin therapy using single or multiple daily insulin injections. A recent meta-analysis RCTs showed that closed-loop insulin delivery enabled an improvement in TIR of 337 min per 24 h (Hedges g = 1.22%, 95% CI 0.84%–1.6%, P < 0.01), with a reduction in time spent in hyperglycemia (time above range [TAR]), and no difference in time spent in hypoglycemia (time below range [TBR]) (20).

The study objective was to assess whether a hybrid closed-loop device (DBLG1) will allow better glycemic control than CSII + CGM in individuals with type 2 diabetes previously treated with CSII in their usual living conditions.

Design

We conducted an interventional, randomized, controlled, cross-over, open-label, multicenter, 13-week study. A relatively small number of individuals with type 2 diabetes benefit from insulin therapy administrated by CSII. For a first study in this population, a crossover study was chosen to achieve a greater statistical power with a limited number of patients included. Data were collected from August 2022 to July 2023. The study protocol was approved by the ethics committee (CPP Ouest I, Tours, France, ID-RCB 2020-A03429-30) and is provided in the Supplementary Material. The safety aspects of the trial were overseen by an independent data and safety monitoring board. All individuals who were included signed informed consent forms after receiving verbal and written information with a time of reflection of several days. The study was registered at ClinicalTrials.gov (NCT05369871). The study is reported according to the Consolidated Standards of Reporting Trials (CONSORT) extension for crossover studies guidelines.

Sample

We recruited outpatients with type 2 diabetes monitored in three hospitals in France, including one university hospital. They were previously treated with CSII in routine clinical care via collaboration with a home care service provider (Agiradom, France). In France, CSII therapy can be administered and reimbursed to patients with type 2 diabetes if they do not have a good glycemic control on multidaily injection therapy. These participants lived at home and were autonomous in their daily activities. They were also autonomous in managing their CSII + FGM therapy at home. Participant eligibility was assessed using clinical and treatment data collected by the home care provider. Inclusion was done in each hospital center.

Inclusion criteria were age >18 years, body weight ≤150 kg, type 2 diabetes diagnosed by a diabetologist, treated with an insulin pump for at least 6 months and stable pharmacological treatment of diabetes for at least 6 months, equipped with a CGM or FGM system, total daily insulin dose <160 IU/24 h, and HbA1c <10%.

Criteria for noninclusion were type 1 diabetes, insulin requirements <8 IU/24 h, a disease that could impact on the physiology of diabetes (i.e., involving interactions with glucose or insulin that could interfere with the medical device, such as corticosteroid treatment), severe uncorrected hearing, visual acuity problems, pancreatectomy, severely impaired pancreas function, renal failure with clearance <30 mL/min/m2, impaired hypoglycemia perception, highly unstable diabetes (patient experiencing problematic hypoglycemia despite up-to-date and appropriate diabetes management), pancreas or islet transplantation, severe neuropathy associated with HbA1c >9%, or severe proliferative retinopathy/maculopathy associated with HbA1c >9% (because of the risk of worsening in the event of rapid correction of glycemic control).

Primary End Point

The primary end point was the difference in percentage TIR [70–180] mg/dL measured using the Dexcom G6 model CGM sensor (Dexcom, San Diego, CA) between 6 weeks of hybrid closed-loop therapy (C for “closed”) and 6 weeks of CSII + CGM (O for “open”). The analysis was performed on data from the last 3 weeks of each 6-week condition.

Secondary End Points

The secondary end points were the differences hybrid closed-loop therapy and CSII + CGM for the following variables:

  • daily glycemic variability measured by the coefficient of variation and SD of glycemia,

  • hyperglycemia measured by the percentage TAR (>180 mg/dL),

  • hypoglycemia measured by the percentage TBR (<70 mg/dL),

  • average daily insulin dose,

  • glucose management indicator (GMI) corresponding to estimated HbA1c,

  • daily physical activity measured in MET by 1-week actimetry,

  • sleep duration and sleep fragmentation measured by 1-week actimetry,

  • satisfaction with diabetes treatment measured by the Diabetes Treatment Satisfaction Questionnaire (DTSQs),

  • treatment safety (prospective collection of severe adverse events), and

  • satisfaction with the therapy system assessed using a usability questionnaire.

To apply the recent recommendations (21) published after the end of the study concerning the reporting of results in studies involving CGM data, we analyzed the following variables that were not prespecified in the protocol:

  • TBR (very low glucose or level 2 hypoglycemia), that is, <54 mg/dL

  • TAR (very high glucose or level 2 hyperglycemia), that is, >250 mg/dL

  • Mean sensor glucose (mg/dL)

Hybrid Closed-Loop System

We used the DBLG1 system, which is composed of an Accu-Chek Insight insulin pump (Roche, Basel, Switzerland) and the DEXCOMG6 CGM system. Transmission is via Bluetooth Low Energy technology and the system uses the Diabeloop application (Regulation v2017.04.20; Diabeloop, Paris, France) installed on an android smartphone (Motorola Moto E XT1524, Motorola, Chicago, IL). The data were computed and accessible in real-time to participants and physicians through the YourLoops web platform (Diabeloop, Paris, France). DBLG1 is a hybrid loop algorithm on which the patient needs to input meals by the amount of carbohydrates ingested or by selecting meal size (small, medium, or large). The physical activity also needs to be declared. Previous work has shown that the most frequent adjustments to the algorithm parameters were generally made within the first 2 weeks, and that glucose measurements were stable after 1 or 2 weeks of active hybrid closed loop (22). We therefore chose to analyze the data after the adjustments to the algorithm settings during the first 2 weeks.

CSII + CGM System

During the CSII + CGM period, participants used their usual insulin pump therapy. They were equipped with a DexcomG6 CGM and had access to continuous glucose data. The use of “hypoglycemia” and “hyperglycemia” alarms was set at the same thresholds for all participants, with an hypoglycemia alarm set at 60 mg/dL for at least 25 min and an hyperglycemia alarm set at 320 mg/dL for at least 20 min.

Study Schedule

The study schedule is shown in Supplementary Fig. 1. Details of each visit are provided in the Supplementary Appendix.

During the closed-loop period, patients began treatment with the new CSII system and the YourLoops software. To enable them to adapt to the new equipment in the event of unexpected shutdown of the closed-loop at a later date, they were left for 7 days with the new CSII system and the loop open, after which the loop was closed. The patient received a call from the nurse on day (D) 1 and D2 when the loop was closed, and then once a week. The doctor looked at the data at D1, D2, D7, and D14 to adjust the algorithm parameters if necessary. The doctor did not intervene in the settings for the following 3 weeks. For the CSII + CGM control period, participants continued their treatment with their usual CSII system, with weekly contacts with a nurse and data visualization on YourLoops at D1, D2, D7, and D14 by the doctor to adjust pump parameters if necessary.

Randomization

Randomization was performed electronically via eCRF, according to a randomization list preestablished by the statistician. The randomization list was stratified by center, by block size 2, using the “proc plan” function of SAS 9.4 software (SAS institute, Cary, NC). The investigator and patient were open about the arm allocated to the patient (O/C or C/O).

Statistical Analysis

We were unable to perform a formal sample size calculation because of lack of relevant data in the literature at the time of study conception. Therefore, the number of people to be included was chosen according to inclusion capacity of each of the three centers, which was estimated at 30 individuals. We anticipated a 30% refusal to participate in the study; therefore, the number of people to be included was set at 20.

Qualitative variables are described by number and percentage. Quantitative variables are reported by means and SDs and differences between periods by mean and 95% CI if the data were normally distributed. Quantitative variables and differences between periods are reported by median and interquartile range (IQR) if the data were not normally distributed. The number and percentage of missing data are reported.

The populations to be analyzed were defined as follows: the modified intention-to-treat (mITT) population was defined as all participants exposed to the second period for at least 24 h, in each arm. The per-protocol population corresponded to the mITT population with no major protocol deviations. A major protocol deviation was defined as stopping the closed-loop for >75% of the planned closed-loop time or completely stopping the planned treatment (closed loop or CSII + CGM treatment). The per-protocol and mITT populations were identical; therefore, only mITT analyses are presented.

The effect of the hybrid closed-loop versus the CSII + CGM on the primary and secondary end points of the study was investigated using specific crossover analyses. These analyses allowed us to study the treatment effect, the order effect, and to control for a possible order × treatment interaction. A significant interaction would mean that the treatment effect depended on the order of administration of the conditions studied. In such cases, only the first period of the crossover was analyzed.

The treatment effect was analyzed by a Student t test if the distribution of interperiod difference values was normally distributed or by a Mann-Whitney test if the distribution of interperiod difference values was not normally distributed. The distribution was assessed visually and using the Shapiro-Wilk test. The comparison was adjusted for treatment order.

The treatment × center interaction was tested on the primary end point using a mixed model with the following factors: order, treatment, center, center × treatment (fixed factors), and subject factor nested within group factor (random factors).

All analyses were performed using SAS 9.4 statistical software (SAS institute, Cary, NC). The threshold of significance was set at 0.05.

Role of the Funding Source

The funding source, DIABELOOP, SA, participated in the study design. It did not participate in data acquisition, statistical analyses, results interpretation, or deciding to submit the manuscript. It did not write the report, except for Fig. 2, which was featured by a member of DIABELOOP.

The study screened 77 individuals, and 20 were included. Three individuals (15%) withdrew (one due to “anxiety about new equipment,” another due to “constraints related to the study and the closed-loop system,” and the last due to “family event”), so 17 individuals were analyzed. The study flowchart is presented in Supplementary Fig. 2. Participant characteristics are presented in Table 1. Mean age of the 17 participants included in the mITT was 63 (SD 9) years, and 64.7% were men. Mean time since diagnosis of diabetes was 24 (SD 9) years and mean treatment time with CSII was 7 (SD 3) years. Mean baseline HbA1c was 7.9% (SD 0.9) or 63 (SD 7) mmol/mol, and mean baseline TIR was 58.9% (SD 20.1). All participants but one used the “small/medium/large meal” function to quantify food intake during the hybrid closed-loop period. One participant used functional insulin therapy to calculate meal bolus doses. The percentage of time spent with active hybrid closed-loop system during the “closed-loop” period was 98% (IQR 95–99) overall, with 99.0% (IQR 97.5–99.0) for the C/O sequence and 97.0% (IQR 89.0–98.0) for the O/C sequence (P = 0.026).

Table 1

Baseline participant characteristics

All participants (N = 17)O/C sequence (n = 9)C/O sequence (n = 8)
Mean (SD) or median (IQR) or n (%)Min; MaxMean (SD) or median (IQR) or n (%)Min; MaxMean (SD) or median (IQR) or n (%)Min; Max
Age (years) 63 (9) 47; 79 63 (6) 53; 75 63 (11) 47; 79 
Male sex, n (%) 11 (64.7)  7 (77.8)  4 (50)  
Height (cm) 172 (9) 155; 190 174 (7) 160; 181 170 (11) 155; 190 
Weight (kg) 97 (16) 73; 121 101 (16) 76; 121 91 (15) 73; 120 
BMI (kg/m²) 32.0 (4.0) 25.0; 40.0 32.9 (4.8) 25.0; 40.0 31.0 (3.0) 28.0; 37.0 
Systolic blood pressure (mmHg) 145 (122–149) 112; 180 136 (125–154) 120; 180 145 (115–149) 112; 162 
Diastolic blood pressure (mmHg) 70 (65–80) 55; 125 75 (69–81) 67; 125 65 (58–80) 55; 93 
Type 2 diabetes characteristics       
 HbA1c (%) 7.9 (0.9) 6.7; 9.7 7.8 (0.7) 6.7; 8.8 8.0 (1.1) 6.8; 9.7 
 HbA1c (mmol/mol) 63 (7) 50; 83 62 (6) 50; 73 64 (9) 51; 83 
 Diabetes duration (years) 24 (9) 13; 47 21 (6) 13; 32 28 (11) 16; 47 
 Duration of insulin pump treatment (years) 7 (3) 1; 15 5 (3) 1; 8 8 (4) 5; 15 
Insulin pump therapy       
 Glycemic target (mg/dL) 110 (110–110) 100; 150 110 (100–110) 100; 120 110 (110–125) 100; 150 
 No. of bolus/day 3.2 (1.8) 0.3; 5.4 3.2 (1.7) 0.3; 5.2 3.3 (2.0) 0.3; 5.4 
 Daily bolus insulin (IU/day) 37.6 (28.6) 0.6; 90.5 39.5 (31.4) 4.2; 90.5 35.6 (26.9) 0.6; 69.6 
 Daily basal insulin (IU/day) 40.4 (14.5) 11.8; 68.6 45.3 (15.8) 19.7; 68.6 34.9 (11.4) 11.8; 48.4 
 Daily total insulin (IU/day) 78.1 (35.9) 23.9; 156.2 84.8 (41.6) 23.9; 156.2 70.5 (29.2) 28.2; 111.2 
CGM       
 TIR, 70–180 mg/dL (%) 58.9 (20.1) 18.0; 86.0 55.5 (18.3) 31.7; 86.0 63.2 (23.0) 18.0; 85.4 
 TAR, >180 mg/dL (%) 40.4 (20.4) 13.8; 81.5 44.1 (18.3) 14.0; 68.3 35.6 (23.4) 13.8; 81.5 
 TBR, <70 mg/dL (%) 0.5 (0.0–0.9) 0.0; 3.5 0.0 (0.0–0.5) 0.0; 0.9 0.8 (0.5–1.9) 0.5; 3.5 
 Mean daily variation coefficient (%) 29.8 (3.8) 21.1; 34.4 28.6 (4.2) 21.1; 32.6 31.4 (2.7) 27.2; 34.4 
 Mean daily SD (g/L) 0.54 (0.10) 0.37; 0.71 0.53 (0.10) 0.37; 0.66 0.54 (0.12) 0.38; 0.71 
 Mean daily GMI (%) 7.7 (1.0) 6.5; 10.1 7.9 (0.9) 6.8; 9.3 7.5 (1.3) 6.5; 10.1 
 Mean daily CGM use (% of 24 h) 81.1 (67.0–91.0) 25.3; 100.0 79.0 (58.3–88.0) 30.6; 97.6 83.3 (75.8–94.1) 25.3; 100.0 
Other antidiabetes medications       
 Metformin, n (%) 11 (64.7)  8 (88.9)  3 (37.5)  
 GLP-1 agonists, n (%) 7 (41.2)  4 (44.4)  3 (37.5)  
 SGLT2 inhibitors (yes), n (%) 5 (29.4)  3 (33.3)  2 (25.0)  
Diabetes complications       
 No. diabetes-related hospitalization during the last year 0.0 (0.0-0.0) 0.0; 1.0 0.0 (0.0-0.0) 0.0; 0.0 0.0 (0.0-0.0) 0.0; 1.0 
 Retinopathy, n (%) 6 (35.3)  4 (44.4)  2 (25.0)  
 Nephropathy, n (%) 6 (35.3)  4 (44.4)  2 (25.0)  
 If yes, dialysis, n (%) 0 (0)  0 (0)  0 (0)  
Diabetic foot injury, n (%) 2 (11.8)  1 (11.1)  1 (12.5)  
Cardiovascular history, n (%) 10 (58.8)  5 (55.6)  5 (62.5)  
 Heart attack 3 (17.6)  2 (22.2)  1 (12.5)  
 Cerebrovascular accident 0 (0)  0 (0)  0 (0)  
 Hypertension 6 (35.3)  3 (33.3)  3 (37.5)  
 Heart failure 0 (0)  0 (0)  0 (0)  
 Other 1 (5.9)  0 (0)  1 (12.5)  
Other medical history, n (%)        
 Depressive disorders 4 (23.5)  1 (11.1)  3 (37.5)  
 Liver cirrhosis 1 (5.9)  1 (11.1)  0 (0)  
 Arrhythmia 2 (11.8)  1 (11.1)  1 (12.5)  
 Cancer 2 (11.8)  1 (11.1)  1 (12.5)  
 Other 3 (17.6)  2 (22.2)  1 (12.5)  
 DTSQs       
 Treatment satisfaction scale total 32 (3) 26; 36 32 (3) 28; 36 32 (3) 26; 35 
 Perceived frequency of       
  Hyperglycemia (n/day) 1.0 (1.0–2.0) 0.0; 4.0 1.0 (1.0–3.0) 0.0; 4.0 1.5 (0.5–2.0) 0.0; 4.0 
  Hypoglycemia (n/day) 1.0 (0.0–2.0) 0.0; 5.0 1.0 (0.0–1.0) 0.0; 2.0 1.5 (0.5–3.0) 0.0; 5.0 
All participants (N = 17)O/C sequence (n = 9)C/O sequence (n = 8)
Mean (SD) or median (IQR) or n (%)Min; MaxMean (SD) or median (IQR) or n (%)Min; MaxMean (SD) or median (IQR) or n (%)Min; Max
Age (years) 63 (9) 47; 79 63 (6) 53; 75 63 (11) 47; 79 
Male sex, n (%) 11 (64.7)  7 (77.8)  4 (50)  
Height (cm) 172 (9) 155; 190 174 (7) 160; 181 170 (11) 155; 190 
Weight (kg) 97 (16) 73; 121 101 (16) 76; 121 91 (15) 73; 120 
BMI (kg/m²) 32.0 (4.0) 25.0; 40.0 32.9 (4.8) 25.0; 40.0 31.0 (3.0) 28.0; 37.0 
Systolic blood pressure (mmHg) 145 (122–149) 112; 180 136 (125–154) 120; 180 145 (115–149) 112; 162 
Diastolic blood pressure (mmHg) 70 (65–80) 55; 125 75 (69–81) 67; 125 65 (58–80) 55; 93 
Type 2 diabetes characteristics       
 HbA1c (%) 7.9 (0.9) 6.7; 9.7 7.8 (0.7) 6.7; 8.8 8.0 (1.1) 6.8; 9.7 
 HbA1c (mmol/mol) 63 (7) 50; 83 62 (6) 50; 73 64 (9) 51; 83 
 Diabetes duration (years) 24 (9) 13; 47 21 (6) 13; 32 28 (11) 16; 47 
 Duration of insulin pump treatment (years) 7 (3) 1; 15 5 (3) 1; 8 8 (4) 5; 15 
Insulin pump therapy       
 Glycemic target (mg/dL) 110 (110–110) 100; 150 110 (100–110) 100; 120 110 (110–125) 100; 150 
 No. of bolus/day 3.2 (1.8) 0.3; 5.4 3.2 (1.7) 0.3; 5.2 3.3 (2.0) 0.3; 5.4 
 Daily bolus insulin (IU/day) 37.6 (28.6) 0.6; 90.5 39.5 (31.4) 4.2; 90.5 35.6 (26.9) 0.6; 69.6 
 Daily basal insulin (IU/day) 40.4 (14.5) 11.8; 68.6 45.3 (15.8) 19.7; 68.6 34.9 (11.4) 11.8; 48.4 
 Daily total insulin (IU/day) 78.1 (35.9) 23.9; 156.2 84.8 (41.6) 23.9; 156.2 70.5 (29.2) 28.2; 111.2 
CGM       
 TIR, 70–180 mg/dL (%) 58.9 (20.1) 18.0; 86.0 55.5 (18.3) 31.7; 86.0 63.2 (23.0) 18.0; 85.4 
 TAR, >180 mg/dL (%) 40.4 (20.4) 13.8; 81.5 44.1 (18.3) 14.0; 68.3 35.6 (23.4) 13.8; 81.5 
 TBR, <70 mg/dL (%) 0.5 (0.0–0.9) 0.0; 3.5 0.0 (0.0–0.5) 0.0; 0.9 0.8 (0.5–1.9) 0.5; 3.5 
 Mean daily variation coefficient (%) 29.8 (3.8) 21.1; 34.4 28.6 (4.2) 21.1; 32.6 31.4 (2.7) 27.2; 34.4 
 Mean daily SD (g/L) 0.54 (0.10) 0.37; 0.71 0.53 (0.10) 0.37; 0.66 0.54 (0.12) 0.38; 0.71 
 Mean daily GMI (%) 7.7 (1.0) 6.5; 10.1 7.9 (0.9) 6.8; 9.3 7.5 (1.3) 6.5; 10.1 
 Mean daily CGM use (% of 24 h) 81.1 (67.0–91.0) 25.3; 100.0 79.0 (58.3–88.0) 30.6; 97.6 83.3 (75.8–94.1) 25.3; 100.0 
Other antidiabetes medications       
 Metformin, n (%) 11 (64.7)  8 (88.9)  3 (37.5)  
 GLP-1 agonists, n (%) 7 (41.2)  4 (44.4)  3 (37.5)  
 SGLT2 inhibitors (yes), n (%) 5 (29.4)  3 (33.3)  2 (25.0)  
Diabetes complications       
 No. diabetes-related hospitalization during the last year 0.0 (0.0-0.0) 0.0; 1.0 0.0 (0.0-0.0) 0.0; 0.0 0.0 (0.0-0.0) 0.0; 1.0 
 Retinopathy, n (%) 6 (35.3)  4 (44.4)  2 (25.0)  
 Nephropathy, n (%) 6 (35.3)  4 (44.4)  2 (25.0)  
 If yes, dialysis, n (%) 0 (0)  0 (0)  0 (0)  
Diabetic foot injury, n (%) 2 (11.8)  1 (11.1)  1 (12.5)  
Cardiovascular history, n (%) 10 (58.8)  5 (55.6)  5 (62.5)  
 Heart attack 3 (17.6)  2 (22.2)  1 (12.5)  
 Cerebrovascular accident 0 (0)  0 (0)  0 (0)  
 Hypertension 6 (35.3)  3 (33.3)  3 (37.5)  
 Heart failure 0 (0)  0 (0)  0 (0)  
 Other 1 (5.9)  0 (0)  1 (12.5)  
Other medical history, n (%)        
 Depressive disorders 4 (23.5)  1 (11.1)  3 (37.5)  
 Liver cirrhosis 1 (5.9)  1 (11.1)  0 (0)  
 Arrhythmia 2 (11.8)  1 (11.1)  1 (12.5)  
 Cancer 2 (11.8)  1 (11.1)  1 (12.5)  
 Other 3 (17.6)  2 (22.2)  1 (12.5)  
 DTSQs       
 Treatment satisfaction scale total 32 (3) 26; 36 32 (3) 28; 36 32 (3) 26; 35 
 Perceived frequency of       
  Hyperglycemia (n/day) 1.0 (1.0–2.0) 0.0; 4.0 1.0 (1.0–3.0) 0.0; 4.0 1.5 (0.5–2.0) 0.0; 4.0 
  Hypoglycemia (n/day) 1.0 (0.0–2.0) 0.0; 5.0 1.0 (0.0–1.0) 0.0; 2.0 1.5 (0.5–3.0) 0.0; 5.0 

Data are mean (SD) or median (IQR) for normally and not normally distributed data, respectively, and minimum; maximum for continuous data. Qualitative data are n (%). There was one missing data value for blood pressure and one missing data value for CCM metrics at baseline.

GLP-1, glucagon-like peptide 1; Min, minimum; Max, maximum; SGLT2, sodium–glucose cotransporter 2.

Primary Outcome

Median (IQR) TIR was 61.0% (55.0–70.0) during CSII + CGM and 76.0% (69.0–84.0) during the closed-loop periods; median difference of TIR between the two periods of treatment was 15.0 percentage points (IQR 8.0–22.0; treatment effect P < 0.001) (Table 2). There was no significant order × intervention (P = 0.18) or treatment × center (P = 0.86) interaction. Figure 1 shows the individual results of the participants. Despite the absence of order × intervention interaction, the magnitude of improvement with the closed-loop is visually more marked in the O/C versus C/O arm, suggesting retention of the closed-loop benefit on the CSII + CGM sequence in the C/O arm. To assess the impact on results of excluding the period of adaptation of the algorithm to the participant during the first 15 days, we evaluated the difference between the two periods for the total time spent in hybrid closed-loop compared with the total period under CSII + CGM. The difference between the two treatment modalities remains significant in favor of the hybrid closed-loop, albeit of lesser magnitude (6.2 percentage points [SD 7.4], P = 0.004) (Supplementary Table 1).

Table 2

Main and secondary outcomes for the differences between closed-loop system and CSII with CGM

NClosed loopCSII + CGMDifference of closed loop minus CSII + CGMP value treatment effectStatistical test for treatment effectP value for normality test of the treatment effectP value for interaction test order x intervention
Main outcome         
 TIR 70–180 mg/dL (%) 17 76.0 (69.0–84.0) 61.0 (55.0–70.0) 15.0 (8.0–22.0) <0.001 Mann-Whitney 0.013 0.18 
Secondary outcomes         
 CGM metrics         
  Mean sensor glucose (mg/dL) 17 158.8 ± 17.3 172.2 ± 20.8 −13.2 (−20.8 to −5.6) 0.002 Student 0.12 0.19 
  TAR >180 mg/dL (%) 17 24.0 (16.0–30.0) 38.0 (30.0–45.0) −15.0 (−22.0 to −8.0) <0.001 Mann-Whitney 0.007 0.18 
  Level 2 hyperglycemia, >250 mg/dL 17 2.3 (1.0–5.3) 7.0 (3.7–9.6) −3.3 (−6.9 to 0.7) 0.014 Student 0.71 0.16 
  TBR <70 mg/dL (%) 17 0.1 (0.0–0.4) 0.3 (0.2–1.0) −0.2 (−0.2 to 0.0) 0.13 Mann-Whitney 0.001 0.38 
  Level 2 hypoglycemia, <54 mg/dL 17 0.00 (0.00–0.05) 0.00 (0.00–0.02) 0.00 (−0.02 to 0.03) 0.89 Mann-Whitney <0.001 0.81 
  Variation coefficient (%) 17 23.0 (3.3) 25.1 (2.9) −2.1 (95% CI −3.6 to 0.7) 0.006 Student 0.30 0.83 
  SD (g/L) 17 0.43 (0.11) 0.50 (0.08) −0.07 (95% CI −0.11 to 0.02) 0.005 Student 0.20 0.54 
  GMI (%) 17 7.1 (0.4) 7.4 (0.5) −0.3 (95% CI −0.5 to 0.1) 0.002 Student 0.12 0.16 
  CGM use (% of 24 h) 17 99.0 (96.9–99.0) 95.0 (93.0–98.0) 2.0 (0.0–5.0) 0.016 Mann-Whitney 0.006 0.65 
 Insulin doses         
  Daily total insulin (IU/day) 17 103.7 (76.1–122.8) 78.0 (59.3–95.3) 10.0 (3.3–30.6) 0.003 Mann-Whitney <0.001 0.36 
 DTSQs         
  Treatment satisfaction scale total 17 31.0 (28.0–35.0) 32.0 (28.0–35.0) −1.0 (−6.0 to 4.0) 0.89 Student 0.15 0.43 
  Perceived frequency of         
   Hyperglycemia (n/day) 17 1.0 (1.0–3.0) 3.0 (1.0–5.0) −2.0 (−3.0 to 1.0) 0.045 Student 0.19 0.65 
   Hypoglycemia (n/day) 17 1.0 (0.0–2.0) 1.0 (0.0–2.0) 0.0 (−1.0 to 1.0) 0.84 Student 0.15 0.27 
 Actimetry         
  Mean daily physical activity (METs) 16 1.07 (1.04–1.09) 1.06 (1.05–1.08) 0.01 (−0.01 to 0.02) 0.16 Mann-Whitney 0.044 0.50 
  Mean daily total sleep time (min) 16 369 (64) 371 (72) −2 (−51 to 47) 0.93 Student 0.21 0.64 
  Mean daily sleep fragmentation index 16       0.038 
NClosed loopCSII + CGMDifference of closed loop minus CSII + CGMP value treatment effectStatistical test for treatment effectP value for normality test of the treatment effectP value for interaction test order x intervention
Main outcome         
 TIR 70–180 mg/dL (%) 17 76.0 (69.0–84.0) 61.0 (55.0–70.0) 15.0 (8.0–22.0) <0.001 Mann-Whitney 0.013 0.18 
Secondary outcomes         
 CGM metrics         
  Mean sensor glucose (mg/dL) 17 158.8 ± 17.3 172.2 ± 20.8 −13.2 (−20.8 to −5.6) 0.002 Student 0.12 0.19 
  TAR >180 mg/dL (%) 17 24.0 (16.0–30.0) 38.0 (30.0–45.0) −15.0 (−22.0 to −8.0) <0.001 Mann-Whitney 0.007 0.18 
  Level 2 hyperglycemia, >250 mg/dL 17 2.3 (1.0–5.3) 7.0 (3.7–9.6) −3.3 (−6.9 to 0.7) 0.014 Student 0.71 0.16 
  TBR <70 mg/dL (%) 17 0.1 (0.0–0.4) 0.3 (0.2–1.0) −0.2 (−0.2 to 0.0) 0.13 Mann-Whitney 0.001 0.38 
  Level 2 hypoglycemia, <54 mg/dL 17 0.00 (0.00–0.05) 0.00 (0.00–0.02) 0.00 (−0.02 to 0.03) 0.89 Mann-Whitney <0.001 0.81 
  Variation coefficient (%) 17 23.0 (3.3) 25.1 (2.9) −2.1 (95% CI −3.6 to 0.7) 0.006 Student 0.30 0.83 
  SD (g/L) 17 0.43 (0.11) 0.50 (0.08) −0.07 (95% CI −0.11 to 0.02) 0.005 Student 0.20 0.54 
  GMI (%) 17 7.1 (0.4) 7.4 (0.5) −0.3 (95% CI −0.5 to 0.1) 0.002 Student 0.12 0.16 
  CGM use (% of 24 h) 17 99.0 (96.9–99.0) 95.0 (93.0–98.0) 2.0 (0.0–5.0) 0.016 Mann-Whitney 0.006 0.65 
 Insulin doses         
  Daily total insulin (IU/day) 17 103.7 (76.1–122.8) 78.0 (59.3–95.3) 10.0 (3.3–30.6) 0.003 Mann-Whitney <0.001 0.36 
 DTSQs         
  Treatment satisfaction scale total 17 31.0 (28.0–35.0) 32.0 (28.0–35.0) −1.0 (−6.0 to 4.0) 0.89 Student 0.15 0.43 
  Perceived frequency of         
   Hyperglycemia (n/day) 17 1.0 (1.0–3.0) 3.0 (1.0–5.0) −2.0 (−3.0 to 1.0) 0.045 Student 0.19 0.65 
   Hypoglycemia (n/day) 17 1.0 (0.0–2.0) 1.0 (0.0–2.0) 0.0 (−1.0 to 1.0) 0.84 Student 0.15 0.27 
 Actimetry         
  Mean daily physical activity (METs) 16 1.07 (1.04–1.09) 1.06 (1.05–1.08) 0.01 (−0.01 to 0.02) 0.16 Mann-Whitney 0.044 0.50 
  Mean daily total sleep time (min) 16 369 (64) 371 (72) −2 (−51 to 47) 0.93 Student 0.21 0.64 
  Mean daily sleep fragmentation index 16       0.038 

Results are expressed as means and SD and differences between periods by means and 95% CIs if the data are normally distributed. Quantitative variables and differences between periods are reported by median and interquartile range if the data are not normally distributed. Mean differences represent the means of the individual differences between the two periods of the study. If the differences were normally distributed, a Student test was realized to look for treatment effect; if not, a Mann-Whitney test was realized.

Figure 1

Time in range during the closed-loop and the CSII + CGM periods.

Figure 1

Time in range during the closed-loop and the CSII + CGM periods.

Close modal

Secondary Outcomes

The comparison between CSII + CGM and closed-loop periods for the secondary outcomes is reported in Table 2. TAR was shorter during the closed-loop than in the CSII + CGM periods, with no difference in TBR. The glycemic variability, assessed by the variation coefficient and SD, and GMI were lower during the closed-loop than the CSII + CGM periods. The time of CGM use (percentage of 24 h) was high in both conditions with 99.0% (IQR 96.9–99.0) during closed-loop and 95.0% (IQR 93.0–98.0) during CSII + CGM, mean difference 2.0 percentage points (IQR 0.0–5.0; P = 0.016). The global results of the CGM during the closed-loop and CSII + CGM are shown in Fig. 2.

Figure 2

Glucose control during the closed-loop and the CSII + CGM periods. The shaded areas show the 95% CI.

Figure 2

Glucose control during the closed-loop and the CSII + CGM periods. The shaded areas show the 95% CI.

Close modal

The total daily insulin dose was higher during the closed-loop than during the CSII + CGM periods. Total DTSQs score did not differ between conditions. The perceived frequency of hyperglycemia was lower with the closed-loop, but the perceived frequency of hypoglycemia did not differ between conditions.

The mean daily physical activity and the mean total sleep time did not differ between conditions. The order × treatment interaction was significant for the sleep fragmentation index. We therefore compared the sleep fragmentation index between closed-loop and CSII + CGM for only the first period of the sequence. The Mean sleep fragmentation index was lower with the closed-loop system than with CSII + CGM (18.2 [SD 5.0] vs. 23.6 [SD 4.9], P = 0.047, respectively).

Adverse Events

Adverse events are reported in Supplementary Table 2. One serious adverse event was attributable to the study. It occurred in a participant who presented with severe hyperglycemia that needed the on-call physician intervention. Hyperglycemia was caused by a folded insulin pump cannula while the participant was in the closed-loop period of the study. Under physician monitoring, the remission of the hyperglycemia occurred without hospitalization. Of note, no severe hypoglycemic events occurred. All adverse events and Common Terminology Criteria for Adverse Events (CTCAE) grades are detailed in Supplementary Table 3.

In addition, body weight was measured at baseline and at the end of both treatment periods. There was no change in weight between the beginning and end of the study neither for the entire population nor for each group: C/O or O/C (Supplementary Table 4).

Usability

The device, including insulin pump therapy, CGM sensor, and algorithm terminal, was found to be easy to use and easy to learn by approximately three-quarters of  the participants. The remaining one-quarter of participants found the application difficult to use and two of those three dropped out because of the constraints of the “hybrid closed-loop” device. Detailed responses to the usability questionnaires and comments related to the device are reported in Supplementary Tables 5–7.

The results of this work showed that, in patients with type 2 diabetes already receiving intensive insulin therapy administered with CSII combined with CGM, implementation of a hybrid closed-loop system improved the median TIR by 15.0 percentage points (IQR 8.0–22.0). This improvement was associated with a reduction in TAR, glucose variability, and GMI and with an increase in the total daily insulin dose. Moreover, the improvement in glycemic control was not achieved at the expense of an increase in TBR. In addition, quality of sleep was better in the closed-loop than the CSII + CGM condition because of a reduction in sleep fragmentation.

CSII has been shown to improve glycemic control in people with type 2 diabetes (4,5). In addition, the use of a FGM or a CGM system also improves HbA1c and TIR in people with type 2 diabetes treated with insulin (insulin pump or multiple daily injections) (6,23). In this context, the added value of a hybrid closed-loop to improve an already intensive treatment with CSII + CGM might not have brought any additional benefit. Previous studies showing benefits of closed-loop therapy in type 2 diabetes have focused on particular situations in which glycemic control is jeopardized: perioperative period (16), dialysis (17), under nutritional support (14), or any medical condition leading to insulin treatment during hospitalization (13). In one study under usual living conditions (18), a fully closed-loop system was compared with standard insulin therapy in a single-center RCT with parallel arms in which 28 people without prior insulin treatment were included. The TIR increased in both groups with an additional benefit in the closed-loop arm. As in our study, the TAR range was lower in the closed-loop arm, with no difference in TBR, and the daily insulin doses were higher in the closed-loop arm. Another multicenter RCT with parallel arms (19) included individuals with uncontrolled insulin-treated type 2 diabetes who depended on a home nurse for insulin injections. They were randomized to a hybrid closed-loop intervention group, managed by a home nurse every 3 days combined with 7–10 days of remote medical supervision, compared with usual treatment group managed by a home nurse every day. TIR improved in the closed-loop arm from 34% (SD 21.3) to 63.0% (SD 9.4), with no increase in TBR.

These studies demonstrated that the closed-loop system increased TIR and reduced time in hyperglycemia without increasing the risk of hypoglycemia in people with type 2 diabetes. In addition to monitoring HbA1c levels, use of these new glycemic control metrics, derived from CGM, is now recommended to monitor patients with type 1 diabetes and type 2 diabetes because they more efficiently capture the glucose variability and the risk of hypoglycemia (24–26). TIR and TAR are correlated with occurrence of albuminuria (27), retinopathy (28), and carotid atherosclerosis (29). In our study, the 70% recommended TIR threshold was reached in ∼75% of the participants with the hybrid closed-loop and the glycemic variability decreased (30).

People living with diabetes have poorer quality of sleep than people without diabetes, partly due to greater sleep fragmentation (31,32) caused by nocturnal hypoglycemia, and/or nocturnal hyperglycemia, leading to polyuria and hence nocturia. Hyperglycemia has been linked to an increased arousal index (32) as well as glycemic variability (31). Therefore, a secondary outcome of our study was to objectively determine the effect of the closed-loop system on sleep quality by using actimetry. Despite interaction with the treatment sequence, sleep fragmentation was reduced, suggesting that the hybrid closed-loop system improved sleep quality. These data are exploratory, given the small number of participants. However, this additional positive effect of hybrid closed-loop therapy should be confirmed in longer duration studies.

Our findings must be interpreted in the light of some limitations. First, the inclusion conditions limited the daily insulin dose to 160 IU/24 h and body weight to 150 kg. These criteria were due partly to the size of the insulin pump reservoir and partly to the algorithm, which was designed not to exceed a cumulative dose of 160 IU/24 h. Therefore, individuals with major insulin requirements were not included, limiting the generalizability of our findings. This problem could be addressed by using more concentrated U500 insulin and adapting the algorithm accordingly. Indeed, beyond the practical advantage of reducing the need to refill the insulin pump tank more than once a day, U500 is more effective for the glycemic control of people with high insulin resistance (33).

The second limitation is the relatively short duration of exposure to the closed-loop period of 6 weeks, including 1 week of open treatment with the new CSII and 2 weeks of adjustment of the closed-loop parameters, which requires close medical follow-up. Given the time needed to adapt to the new equipment and treatment modalities, although glycemic control was improved, participants did not necessarily feel the benefit of the hybrid closed-loop, as reflected by the lack of difference in satisfaction between treatment modalities. A longer-term study could provide a better assessment of the impact of this hybrid closed-loop treatment on satisfaction and quality of life, once the learning curve has been overcome. It would also enable a more reliable assessment of safety and efficacy. In addition, although patients were above the target of 70% CGM use time in clinical routine at the start of the study, their participation in the study with the frequent nurse’s calls most likely increased CGM use time from 81 to 95% during the control period and to 99% during the closed-loop period. We were careful to have the same number of patient interactions during both periods, so as not to induce follow-up bias between the two arms. However, the fact that the study was not blinded may explain why CGM time was slightly higher during the closed-loop period, as patients were probably more attentive to their treatment during under hybrid closed-loop, which was new to them. We verified, in a post hoc analysis, that this difference had no impact to the results for TIR (Pearson correlation between TIR and time with active CGM on the whole sample was r = −0.18, P = 0.49).

Finally, the relatively small number of patients (n = 17) who completed the study, and the use of CSII + CGM as a comparator, to assess the benefit of a hybrid closed-loop in patients with type 2 diabetes, limits the generalizability of the results; indeed, CSII treatment is not generally a reimbursed treatment outside France. Our study should therefore be seen as a proof of concept that highlights which insulin delivery modality could provide the best glycemic control in patients with type 2 diabetes.

In summary, this study showed that a hybrid closed-loop therapy further improves glycemic control in people with type 2 diabetes already optimally treated with CSII + CGM. Long-term studies are now required to measure the benefits of closed-loop therapy on long-term glycemic control, quality of life, treatment satisfaction, and prevention of diabetes-related complications.

Clinical trial reg. no. NCT05369871, ClinicalTrials.gov

This article contains supplementary material online at https://doi.org/10.2337/figshare.26190590.

Acknowledgments. The authors thank Johanna Robertson (https://johannarobertson.fr/) for English editing, who received financial support for her participation. The authors thank Myriam Haddouche, Laure Nasse, and M’Barka Daoukhi, research professionals, and the nurses from Agiràdom, SA, for their help during the study.

Funding. This study was funded by DIABELOOP, Inc. (France), under agreements with the Contract Research Organization ICADOM (France), the CHU Grenoble Alpes, CH Annecy-Genevois, and CH Métropole Savoie.

Duality of Interest. A.-L.B. is married to Jean-Christian Borel, Scientific Director of Contract Research Organization ICADOM (France). C.B., E.J., and N.A. are employees of the Contract Research Organization ICADOM (France). P.-Y.B. has received personal fees as Chief Medical Officer from DIABELOOP SA. A.-L.B. (academic) and P.-Y.B. (academic involved as expert in the scientific board of DIABELOOP, Inc., which sponsored the study) directly accessed and verified the underlying data reported in the manuscript. No other potential conflicts of interest relevant to this article were reported.

Author Contributions. A.-L.B. wrote the manuscript. A.-L.B., S.L., C.W., E.J., C.B., H.A., and P.-Y.B. contributed to data collection. A.-L.B. S.L., C.W., H.A., and P.-Y.B. contributed to the investigation. A.-L.B. S.L., E.J., C.B., and P.-Y.B. contributed to study design and methodology. A.-L.B., S.L., and P.-Y.B. contributed to data interpretation. A.-L.B., N.A., and P.-Y.B. created the figures. A.-L.B. and P.-Y.B. conducted the literature search and conceptualized the study. S.L., C.W., H.A., and P.-Y.B. contributed to review. E.J. and C.B. contributed to data curation and to project administration. N.A. contributed to data analysis. All authors approved the final version of the manuscript. A.-L.B. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Parts of the study were presented as an oral communication at Le congrès de la Société Francophone du Diabète 2024, Toulouse, France, 19–22 March 2024.

Handling Editors. The journal editors responsible for overseeing the review of the manuscript were Cheryl A.M. Anderson and Rodica Pop-Busui.

1.
Magliano
DJ
,
Boyko
E.J
.
IDF Diabetes Atlas.
10th ed.
Brussels
,
2021
.
2.
Tancredi
M
,
Rosengren
A
,
Svensson
A-M
, et al
.
Excess mortality among persons with type 2 diabetes
.
N Engl J Med
2015
;
373
:
1720
1732
3.
Fosse-Edorh
SPC
,
Saboni
L
,
Mandereau-Bruno
L
, et al
.
Études Entred: un dispositif pour améliorer la connaissance de l’état de santé des personnes présentant un diabète en France – Premiers résultats de la troisième édition conduite en métropole en 2019
.
Bull Epidémiol Hebd
2022
;
22
:
383
392
4.
Reznik
Y
,
Cohen
O
,
Aronson
R
, et al;
OpT2mise Study Group
.
Insulin pump treatment compared with multiple daily injections for treatment of type 2 diabetes (OpT2mise): a randomised open-label controlled trial
.
Lancet
2014
;
384
:
1265
1272
5.
Bétry
C
,
Lablanche
S
,
Carvalho
M
, et al
.
Effect of a lifestyle intervention to prevent weight gain at initiation of insulin pump therapy in type 2 diabetes: A randomized, controlled, multicentre trial
.
Diabetes Res Clin Pract
2023
;
200
:
110698
6.
Uhl
S
,
Choure
A
,
Rouse
B
,
Loblack
A
,
Reaven
P
.
Effectiveness of continuous glucose monitoring on metrics of glycemic control in type 2 diabetes mellitus: a systematic review and meta-analysis of randomized controlled trials
.
J Clin Endocrinol Metab
2024
;
109
:
1119
1131
7.
Tauschmann
M
,
Thabit
H
,
Bally
L
, et al.
APCam11 Consortium
.
Closed-loop insulin delivery in suboptimally controlled type 1 diabetes: a multicentre, 12-week randomised trial
.
Lancet
2018
;
392
:
1321
1329
8.
Brown
SA
,
Kovatchev
BP
,
Raghinaru
D
, et al;
iDCL Trial Research Group
.
Six-month randomized, multicenter trial of closed-loop control in type 1 diabetes
.
N Engl J Med
2019
;
381
:
1707
1717
9.
Brown
SA
,
Forlenza
GP
,
Bode
BW
, et al;
Omnipod 5 Research G
.
Multicenter trial of a tubeless, on-body automated insulin delivery system with customizable glycemic targets in pediatric and adult participants with type 1 diabetes
.
Diabetes Care
2021
;
44
:
1630
1640
10.
Carlson
AL
,
Sherr
JL
,
Shulman
DI
, et al
.
Safety and glycemic outcomes during the MiniMed™ advanced hybrid closed-loop system pivotal trial in adolescents and adults with type 1 diabetes
.
Diabetes Technol Ther
2022
;
24
:
178
189
11.
Benhamou
P-Y
,
Franc
S
,
Reznik
Y
, et al.
I DIABELOOP WP7 Trial Investigators
.
Closed-loop insulin delivery in adults with type 1 diabetes in real-life conditions: a 12-week multicentre, open-label randomised controlled crossover trial
.
Lancet Digit Health
2019
;
1
:
e17
e25
12.
Thabit
H
,
Hartnell
S
,
Allen
JM
, et al
.
Closed-loop insulin delivery in inpatients with type 2 diabetes: a randomised, parallel-group trial
.
Lancet Diabetes Endocrinol
2017
;
5
:
117
124
13.
Bally
L
,
Thabit
H
,
Hartnell
S
, et al
.
Closed-loop insulin delivery for glycemic control in noncritical care
.
N Engl J Med
2018
;
379
:
547
556
14.
Boughton
CK
,
Bally
L
,
Martignoni
F
, et al
.
Fully closed-loop insulin delivery in inpatients receiving nutritional support: a two-centre, open-label, randomised controlled trial
.
Lancet Diabetes Endocrinol
2019
;
7
:
368
377
15.
Kumareswaran
K
,
Thabit
H
,
Leelarathna
L
, et al
.
Feasibility of closed-loop insulin delivery in type 2 diabetes: a randomized controlled study
.
Diabetes Care
2014
;
37
:
1198
1203
16.
Herzig
D
,
Suhner
S
,
Roos
J
, et al
.
Perioperative fully closed-loop insulin delivery in patients undergoing elective surgery: an open-label, randomized controlled trial
.
Diabetes Care
2022
;
45
:
2076
2083
17.
Boughton
CK
,
Tripyla
A
,
Hartnell
S
, et al
.
Fully automated closed-loop glucose control compared with standard insulin therapy in adults with type 2 diabetes requiring dialysis: an open-label, randomized crossover trial
.
Nat Med
2021
;
27
:
1471
1476
18.
Daly
AB
,
Boughton
CK
,
Nwokolo
M
, et al
.
Fully automated closed-loop insulin delivery in adults with type 2 diabetes: an open-label, single-center, randomized crossover trial
.
Nat Med
2023
;
29
:
203
208
19.
Reznik
Y
,
Carvalho
M
,
Fendri
S
, et al
.
Should people with type 2 diabetes treated by multiple daily insulin injections with home health care support be switched to hybrid closed-loop? The CLOSE AP+ randomized controlled trial
.
Diabetes Obes Metab
2024
;
26
:
622
630
20.
Amer
BE
,
Yaqout
YE
,
Abozaid
AM
,
Afifi
E
,
Aboelkhier
MM
.
Does fully closed-loop automated insulin delivery improve glycaemic control in patients with type 2 diabetes? A meta-analysis of randomized controlled trials
.
Diabet Med
2023
;
41
:
e15196
21.
Battelino
T
,
Alexander
CM
,
Amiel
SA
, et al
.
Continuous glucose monitoring and metrics for clinical trials: an international consensus statement
.
Lancet Diabetes Endocrinol
2023
;
11
:
42
57
22.
Benhamou
PY
,
Madrolle
S
,
Lablanche
S
, et al
.
Comment on Leelarathna et al. Duration of hybrid closed-loop insulin therapy to achieve representative glycemic outcomes in adults with type 1 diabetes. Diabetes Care
2020
;
43
:
e38
e39
.
Diabetes Care
2020;43:e167
23.
Haak
T
,
Hanaire
H
,
Ajjan
R
,
Hermanns
N
,
Riveline
J-P
,
Rayman
G
.
Use of flash glucose-sensing technology for 12 months as a replacement for blood glucose monitoring in insulin-treated type 2 diabetes
.
Diabetes Ther
2017
;
8
:
573
586
24.
Advani
A
.
Positioning time in range in diabetes management
.
Diabetologia
2020
;
63
:
242
252
25.
Davies
MJ
,
Aroda
VR
,
Collins
BS
, et al
.
Management of hyperglycemia in type 2 diabetes, 2022. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD)
.
Diabetes Care
2022
;
45
:
2753
2786
26.
Holt
RIG
,
DeVries
JH
,
Hess-Fischl
A
, et al
.
The management of type 1 diabetes in adults. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD)
.
Diabetes Care
2021
;
44
:
2589
2625
27.
Yoo
JH
,
Choi
MS
,
Ahn
J
, et al
.
Association between continuous glucose monitoring-derived time in range, other core metrics, and albuminuria in type 2 diabetes
.
Diabetes Technol Ther
2020
;
22
:
768
776
28.
Lu
J
,
Ma
X
,
Zhou
J
, et al
.
Association of time in range, as assessed by continuous glucose monitoring, with diabetic retinopathy in type 2 diabetes
.
Diabetes Care
2018
;
41
:
2370
2376
29.
Mita
T
,
Katakami
N
,
Okada
Y
, et al
.
Continuous glucose monitoring-derived time in range and CV are associated with altered tissue characteristics of the carotid artery wall in people with type 2 diabetes
.
Diabetologia
2023
;
66
:
2356
2367
30.
Battelino
T
,
Danne
T
,
Bergenstal
RM
, et al
.
Clinical targets for continuous glucose monitoring data interpretation: recommendations from the international consensus on time in range
.
Diabetes Care
2019
;
42
:
1593
1603
31.
Brandt
R
,
Park
M
,
Wroblewski
K
,
Quinn
L
,
Tasali
E
,
Cinar
A
.
Sleep quality and glycaemic variability in a real-life setting in adults with type 1 diabetes
.
Diabetologia
2021
;
64
:
2159
2169
32.
Griggs
S
,
Redeker
NS
,
Jeon
S
,
Grey
M
.
Daily variations in sleep and glucose in adolescents with type 1 diabetes
.
Pediatr Diabetes
2020
;
21
:
1493
1501
33.
Deberles
E
,
Morello
R
,
Hardouin
J
, et al
.
The switch from rapid-acting to concentrated regular insulin improves glucose control in type 2 diabetes patients on pump therapy: a cohort survey
.
Diabetes Metab
2022
;
48
:
101300
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.