Improvements in Glycemic Control Achieved by Altering the tmax Setting in the iLet® Bionic Pancreas When Using Fast-Acting Insulin Aspart: A Randomized Trial

Steven J Russell, Courtney Balliro, Magnus Ekelund, Firas El-Khatib, Tina Graungaard, Evelyn Greaux, Mallory Hillard, Rabab Z Jafri, Naveen Rathor, Raj Selagamsetty, Jordan Sherwood, Edward R Damiano, Steven J Russell, Courtney Balliro, Magnus Ekelund, Firas El-Khatib, Tina Graungaard, Evelyn Greaux, Mallory Hillard, Rabab Z Jafri, Naveen Rathor, Raj Selagamsetty, Jordan Sherwood, Edward R Damiano

Abstract

Introduction: We investigated the safety of, and glucose control by, the insulin-only configuration of the iLet® bionic pancreas delivering fast-acting insulin aspart (faster aspart), using the same insulin-dosing algorithm but different time to maximal serum drug concentration (tmax) settings, in adults with type 1 diabetes.

Methods: We performed a single-center, single-blinded, crossover (two 7-day treatment periods) escalation trial over three sequential cohorts. Participants from each cohort were randomized to a default tmax setting (t65 [tmax = 65 min]) followed by a non-default tmax setting (t50 [tmax = 50 min; cohort 1], t40 [tmax = 40 min; cohort 2], t30 [tmax = 30 min; cohort 3]), or vice versa, all with faster aspart. Each cohort randomized eight new participants if escalation-stopping criteria were not met in the previous cohort.

Results: Overall, 24 participants were randomized into three cohorts. Two participants discontinued treatment, one due to reported 'low blood glucose' during the first treatment period of cohort 3 (t30). Mean time in low sensor glucose (< 54 mg/dl, primary endpoint) was < 1.0% for all tmax settings. Mean sensor glucose in cohorts 1 and 2 was significantly lower at non-default versus default tmax settings, with comparable insulin dosing. The mean time sensor glucose was in range (70-180 mg/dl) was > 70% for all cohorts, except the default tmax setting in cohort 1. No severe hypoglycemic episodes were reported. Furthermore, there were no clinically significant differences in adverse events between the groups.

Conclusion: There were no safety concerns with faster aspart in the iLet at non-default tmax settings. Improvements were observed in mean sensor glucose without increases in low sensor glucose at non-default tmax settings.

Trial registration: ClinicalTrials.gov, NCT03816761.

Keywords: Algorithm; Automated delivery; Bionic pancreas; Faster aspart; Glucose control; Insulin; Type 1 diabetes.

Figures

Fig. 1
Fig. 1
Trial design. *During the ‘in-patient’ period, participants stayed in a hotel close to the hospital. Participants were permitted to leave the hotel during the day; however, they informed the on-call study provider of their whereabouts. The ‘out-patient’ period was defined as the period after the participant had been discharged to home until the end of treatment. †Continuation to the next cohort only occurred if the following criteria did not occur in ≥ 2 subjects on the same tmax setting within a cohort: time in low sensor glucose > 2% of the total time on treatment; ≥ 1 treatment-emergent severe hypoglycemic episode. tmax, time to maximal serum drug concentration
Fig. 2
Fig. 2
Time spent in low sensor glucose (a cohort 1, b cohort 2, and c cohort 3, and mean sensor glucose for d cohort 1, e cohort 2, and f cohort 3. Time spent in low sensor glucose was calculated as the percentage of available sensor glucose values below the threshold. Each blue line represents the sensor glucose profile for an individual participant in their respective cohort; different shades indicate different tmax settings. Yellow lines indicate mean over participants in a cohort. The blue dot represents the participant that discontinued treatment during the first treatment period, i.e., the participant has no assessment on the default (t65) setting. tmax, time to maximal serum drug concentration

References

    1. Nimri R, Nir J, Phillip M. Insulin pump therapy. Am J Ther. 2020;27:e30–e41. doi: 10.1097/MJT.0000000000001097.
    1. Yeh HC, Brown TT, Maruthur N, et al. Comparative effectiveness and safety of methods of insulin delivery and glucose monitoring for diabetes mellitus: a systematic review and meta-analysis. Ann Intern Med. 2012;157:336–347. doi: 10.7326/0003-4819-157-5-201209040-00508.
    1. Misso ML, Egberts KJ, Page M, O'Connor D, Shaw J. Continuous subcutaneous insulin infusion (CSII) versus multiple insulin injections for type 1 diabetes mellitus. Cochrane Database Syst Rev. 2010 doi: 10.1002/14651858.CD005103.pub2.
    1. REPOSE Study Group Relative effectiveness of insulin pump treatment over multiple daily injections and structured education during flexible intensive insulin treatment for type 1 diabetes: cluster randomised trial (REPOSE) BMJ. 2017;356:j1285.
    1. Foster NC, Beck RW, Miller KM, et al. State of type 1 diabetes management and outcomes from the T1D exchange in 2016–2018. Diabetes Technol Ther. 2019;21:66–72. doi: 10.1089/dia.2018.0384.
    1. van den Boom L, Karges B, Auzanneau M, et al. Temporal trends and contemporary use of insulin pump therapy and glucose monitoring among children, adolescents, and adults with type 1 diabetes between 1995 and 2017. Diabetes Care. 2019;42:2050–2056. doi: 10.2337/dc19-0345.
    1. Pettus JH, Zhou FL, Shepherd L, et al. Incidences of severe hypoglycemia and diabetic ketoacidosis and prevalence of microvascular complications stratified by age and glycemic control in U.S. adult patients with type 1 diabetes: a real-world study. Diabetes Care. 2019;42:2220–2227. doi: 10.2337/dc19-0830.
    1. Pathak V, Pathak NM, O'Neill CL, Guduric-Fuchs J, Medina RJ. Therapies for type 1 diabetes: current scenario and future perspectives. Clin Med Insights Endocrinol Diabetes. 2019;12:1179551419844521. doi: 10.1177/1179551419844521.
    1. El-Khatib FH, Russell SJ, Magyar KL, et al. Autonomous and continuous adaptation of a bihormonal bionic pancreas in adults and adolescents with type 1 diabetes. J Clin Endocrinol Metab. 2014;99:1701–1711. doi: 10.1210/jc.2013-4151.
    1. El-Khatib FH, Russell SJ, Nathan DM, Sutherlin RG, Damiano ER. A bihormonal closed-loop artificial pancreas for type 1 diabetes. Sci Transl Med. 2010;2:27ra. doi: 10.1126/scitranslmed.3000619.
    1. El-Khatib FH, Balliro C, Hillard MA, et al. Home use of a bihormonal bionic pancreas versus insulin pump therapy in adults with type 1 diabetes: a multicentre randomised crossover trial. Lancet. 2017;389:369–380. doi: 10.1016/S0140-6736(16)32567-3.
    1. Russell SJ, Hillard MA, Balliro C, et al. Day and night glycaemic control with a bionic pancreas versus conventional insulin pump therapy in preadolescent children with type 1 diabetes: a randomised crossover trial. Lancet Diabetes Endocrinol. 2016;4:233–243. doi: 10.1016/S2213-8587(15)00489-1.
    1. Kildegaard J, Buckley ST, Nielsen RH, et al. Elucidating the mechanism of absorption of fast-acting insulin aspart: the role of niacinamide. Pharm Res. 2019;36:49. doi: 10.1007/s11095-019-2578-7.
    1. Heise T, Pieber TR, Danne T, Erichsen L, Haahr H. A pooled analysis of clinical pharmacology trials investigating the pharmacokinetic and pharmacodynamic characteristics of fast-acting insulin aspart in adults with type 1 diabetes. Clin Pharmacokinet. 2017;56:551–559. doi: 10.1007/s40262-017-0514-8.
    1. Heise T, Zijlstra E, Nosek L, Rikte T, Haahr H. Pharmacological properties of faster-acting insulin aspart vs insulin aspart in patients with type 1 diabetes receiving continuous subcutaneous insulin infusion: a randomized, double-blind, crossover trial. Diabetes Obes Metab. 2017;19:208–215. doi: 10.1111/dom.12803.
    1. Klonoff DC, Evans ML, Lane W, et al. A randomized, multicentre trial evaluating the efficacy and safety of fast-acting insulin aspart in continuous subcutaneous insulin infusion in adults with type 1 diabetes (onset 5) Diabetes Obes Metab. 2019;21:961–967. doi: 10.1111/dom.13610.
    1. Bode BW, Johnson JA, Hyveled L, Tamer SC, Demissie M. Improved postprandial glycemic control with faster-acting insulin aspart in patients with type 1 diabetes using continuous subcutaneous insulin infusion. Diabetes Technol Ther. 2017;19:25–33. doi: 10.1089/dia.2016.0350.
    1. Zijlstra E, Demissie M, Graungaard T, Heise T, Nosek L, Bode B. Investigation of pump compatibility of fast-acting insulin aspart in subjects with type 1 diabetes. J Diabetes Sci Technol. 2018;12:145–151. doi: 10.1177/1932296817730375.
    1. Dovc K, Piona C, Yesiltepe Mutlu G, et al. Faster compared with standard insulin aspart during day-and-night fully closed-loop insulin therapy in type 1 diabetes: a double-blind randomized crossover trial. Diabetes Care. 2020;43:29–36. doi: 10.2337/dc19-0895.
    1. World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310:2191–2194. doi: 10.1001/jama.2013.281053.
    1. The International Council for Harmonisation (ICH). ICH Harmonised Tripartite Guideline. Guideline for good clinical practice E6(R2), current step 4 version. 2016. . Accessed Feb 01, 2021.
    1. US Food and Drug Administration. Electronic code of federal regulations. 2021. . Accessed Feb, 01 2021.
    1. Seaquist ER, Anderson J, Childs B, et al. Hypoglycemia and diabetes: a report of a workgroup of the American Diabetes Association and the Endocrine Society. Diabetes Care. 2013;36:1384–1395. doi: 10.2337/dc12-2480.
    1. Hsu L, Buckingham B, Basina M, et al. Fast-acting insulin aspart use with the minimed(TM) 670G system. Diabetes Technol Ther. 2021;23:1–7. doi: 10.1089/dia.2020.0083.
    1. Herzig D, Dehais J, Prost JC, et al. Pharmacokinetics of faster and standard insulin aspart during fully closed-loop insulin delivery in type 2 diabetes. Diabetes Technol Ther. 2020;22:691–696. doi: 10.1089/dia.2019.0477.
    1. Bally L, Herzig D, Ruan Y, et al. Short-term fully closed-loop insulin delivery using faster insulin aspart compared with standard insulin aspart in type 2 diabetes. Diabetes Obes Metab. 2019;21:2718–2722. doi: 10.1111/dom.13861.
    1. 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. doi: 10.2337/dci19-0028.

Source: PubMed

3
Iratkozz fel