PREVENT: A Randomized, Placebo-controlled Crossover Trial of Avexitide for Treatment of Postbariatric Hypoglycemia

Colleen M Craig, Helen Margaret Lawler, Clare Jung Eun Lee, Marilyn Tan, Dawn Belt Davis, Jenny Tong, Michele Glodowski, Elisa Rogowitz, Rowan Karaman, Tracey L McLaughlin, Lisa Porter, Colleen M Craig, Helen Margaret Lawler, Clare Jung Eun Lee, Marilyn Tan, Dawn Belt Davis, Jenny Tong, Michele Glodowski, Elisa Rogowitz, Rowan Karaman, Tracey L McLaughlin, Lisa Porter

Abstract

Context: Postbariatric hypoglycemia (PBH), characterized by enteroinsular axis overstimulation and hyperinsulinemic hypoglycemia, is a complication of bariatric surgery for which there is no approved therapy.

Objective: To evaluate efficacy and safety of avexitide [exendin (9-39)], a glucagon-like peptide-1 antagonist, for treatment of PBH.

Methods: A multicenter, Phase 2, randomized, placebo-controlled crossover study (PREVENT). Eighteen female patients with PBH were given placebo for 14 days followed by avexitide 30 mg twice daily and 60 mg once daily, each for 14 days in random order. The main outcome measures were glucose nadir and insulin peak during mixed-meal tolerance testing (MMTT) and hypoglycemic events captured by self-monitoring of blood glucose (SMBG), electronic diary, and blinded continuous glucose monitoring (CGM).

Results: Compared with placebo, avexitide 30 mg twice daily and 60 mg once daily raised the glucose nadir by 21% (P = .001) and 26% (P = .0002) and lowered the insulin peak by 23% (P = .029) and 21% (P = .042), corresponding to 50% and 75% fewer participants requiring rescue during MMTT, respectively. Significant reductions in rates of Levels 1 to 3 hypoglycemia were observed, defined, respectively, as SMBG <70 mg/dL, SMBG <54 mg/dL, and a severe event characterized by altered mental and/or physical function requiring assistance. CGM demonstrated reductions in hypoglycemia without induction of clinically relevant hyperglycemia. Avexitide was well tolerated, with no increase in adverse events.

Conclusion: Avexitide administered for 28 days was well tolerated and resulted in robust and consistent improvements across multiple clinical and metabolic parameters, reinforcing the targeted therapeutic approach and demonstrating durability of effect. Avexitide may represent a first promising treatment for patients with severe PBH.

Trial registration: ClinicalTrials.gov NCT03373435.

Keywords: GLP-1 antagonist; PBH; Postbariatric hypoglycemia; avexitide; exendin (939); hyperinsulinemic hypoglycemia.

© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society.

Figures

Figure 1.
Figure 1.
Study Schematic for the PREVENT Trial (above) and mixed meal tolerance test (MMTT) sampling timepoints (below). Avexitide 30 mg twice daily, avexitide 30 mg dose every 12 hours; avexitide 60 mg once daily, avexitide 60 mg dose once each morning. CGM, continuous glucose monitoring; eDiary, electronic diary; SMBG, self-monitoring of blood glucose; SC, subcutaneous.
Figure 2.
Figure 2.
Study profile (Consort). Twenty-two patients were screened and 18 were randomized and completed the study. *Evaluable set was defined as all randomized patients who received at least Treatment Period 1 placebo and Treatment Period 2 active treatment with blood glucose nadir measured during MMTT in both Treatment Periods 1 and 2, without any major protocol deviations that may confound the interpretation of efficacy. One participant was excluded from the Evaluable set analysis because glycemic rescue was not administered as indicated per protocol during the Period 1 placebo MMTT.
Figure 3.
Figure 3.
Mean postprandial plasma glucose (A) and insulin (C) concentrations and mean postprandial plasma glucose nadir (B) and insulin peak (D) in response to MMTT provocation at the end of placebo (grey/dotted line), avexitide 30 mg twice daily (light blue/solid line), and avexitide 60 mg once daily (dark blue/dashed line) treatment periods. Inset within (A) shows an enlargement of axes detailing the glucose values during the 90-180 minute time period. The chart below (A) shows the number of evaluable patients at each sampling timepoint by treatment regimen, representing the number of participants not having required rescue by that timepoint. *P < .05; **P < .01; ***P < .001 for avexitide 30 mg twice daily vs placebo; +P < .05; ++P < .01, and +++P < .001 for avexitide 60 mg once daily vs placebo. Avexitide 30 mg twice daily, avexitide 30 mg dose every 12 hours; avexitide 60 mg once daily, avexitide 60 mg dose once each morning.
Figure 4.
Figure 4.
Mean avexitide plasma concentration by dosing regimen: avexitide 30 mg twice daily; light blue/solid line, and avexitide 60 mg once daily; dark blue/dashed line on Days 29 and 43. On each of Days 29 and 43, a trough PK sample was collected immediately before the T = −90 minute injection of study drug. T = 0 minutes represents the start of Ensure Compact drink consumption. T = 0, 60, and 180 PK samples were collected at the same time as the plasma glucose and hormonal (insulin, C-peptide, GLP-1, glucagon) draws with PK sampling additionally obtained at T = 330 minutes.

References

    1. Ward ZJ, Bleich SN, Cradock AL, et al. . Projected U.S. state-level prevalence of adult obesity and severe obesity. N Engl J Med. 2019;381(25):2440-2450.
    1. Sjöström L, Lindroos AK, Peltonen M, et al. ; Swedish Obese Subjects Study Scientific Group . Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med. 2004;351(26):2683-2693.
    1. Sjöström L, Peltonen M, Jacobson P, et al. . Association of bariatric surgery with long-term remission of type 2 diabetes and with microvascular and macrovascular complications. JAMA 2014;311(22):2297-2304.
    1. Arterburn DE, Johnson E, Coleman KJ, et al. . Weight outcomes of sleeve gastrectomy and gastric bypass compared to nonsurgical treatment. Ann Surg. 2020. Published online March 13, 2020. doi: 10.1097/SLA.0000000000003826
    1. Kwok CS, Pradhan A, Khan MA, et al. . Bariatric surgery and its impact on cardiovascular disease and mortality: a systematic review and meta-analysis. Int J Cardiol. 2014;173(1):20-28.
    1. Adams TD, Gress RE, Smith SC, et al. . Long-term mortality after gastric bypass surgery. N Engl J Med. 2007;357(8):753-761.
    1. American Society for Metabolic and Bariatric Surgery. Estimate of bariatric surgery numbers, 2011-2017. ProMED-mail website. Accessed June 26, 2018.
    1. Rubino F, Nathan DM, Eckel RH, et al. ; Delegates of the 2nd Diabetes Surgery Summit . Metabolic surgery in the treatment algorithm for type 2 diabetes: a joint statement by International Diabetes Organizations. Diabetes Care. 2016;39(6):861-877.
    1. Lee CJ, Brown TT, Schweitzer M, Magnuson T, Clark JM. The incidence and risk factors associated with developing symptoms of hypoglycemia after bariatric surgery. Surg Obes Relat Dis. 2018;14(6):797-802.
    1. Lee CJ, Clark JM, Schweitzer M, et al. . Prevalence of and risk factors for hypoglycemic symptoms after gastric bypass and sleeve gastrectomy. Obesity (Silver Spring). 2015;23(5):1079-1084.
    1. Kefurt R, Langer FB, Schindler K, Shakeri-Leidenmühler S, Ludvik B, Prager G. Hypoglycemia after Roux-En-Y gastric bypass: detection rates of continuous glucose monitoring (CGM) versus mixed meal test. Surg Obes Relat Dis. 2015;11(3):564-569.
    1. Brix JM, Kopp HP, Höllerl F, Schernthaner GH, Ludvik B, Schernthaner G. Frequency of hypoglycaemia after different bariatric surgical procedures. Obes Facts. 2019;12(4):397-406.
    1. Larraufie P, Roberts GP, McGavigan AK, et al. . Important role of the GLP-1 axis for glucose homeostasis after bariatric surgery. Cell Rep. 2019;26(6):1399-1408.e6.
    1. Elliott JA, Docherty NG, Murphy CF, et al. . Changes in gut hormones, glycaemic response and symptoms after oesophagectomy. Br J Surg. 2019;106(6):735-746.
    1. Calabria AC, Charles L, Givler S, De León DD. Postprandial hypoglycemia in children after gastric surgery: clinical characterization and pathophysiology. Horm Res Paediatr. 2016;85(2):140-146.
    1. Kim SH, Liu TC, Abbasi F, et al. . Plasma glucose and insulin regulation is abnormal following gastric bypass surgery with or without neuroglycopenia. Obes Surg. 2009;19(11):1550-1556.
    1. Service GJ, Thompson GB, Service FJ, Andrews JC, Collazo-Clavell ML, Lloyd RV. Hyperinsulinemic hypoglycemia with nesidioblastosis after gastric-bypass surgery. N Engl J Med. 2005;353(3):249-254.
    1. Salehi M, Vella A, McLaughlin T, Patti ME. Hypoglycemia after gastric bypass surgery: current concepts and controversies. J Clin Endocrinol Metab. 2018;103(8):2815-2826.
    1. Patti ME, McMahon G, Mun EC, et al. . Severe hypoglycaemia post-gastric bypass requiring partial pancreatectomy: evidence for inappropriate insulin secretion and pancreatic islet hyperplasia. Diabetologia. 2005;48(11):2236-2240.
    1. Kim SH, Abbasi F, Lamendola C, Reaven GM, McLaughlin T. Glucose-stimulated insulin secretion in gastric bypass patients with hypoglycemic syndrome: no evidence for inappropriate pancreatic beta-cell function. Obes Surg. 2010;20(8):1110-1116.
    1. Salehi M, Prigeon RL, D’Alessio DA. Gastric bypass surgery enhances glucagon-like peptide 1-stimulated postprandial insulin secretion in humans. Diabetes. 2011;60(9):2308-2314.
    1. Korner J, Bessler M, Inabnet W, Taveras C, Holst JJ. Exaggerated glucagon-like peptide-1 and blunted glucose-dependent insulinotropic peptide secretion are associated with Roux-en-Y gastric bypass but not adjustable gastric banding. Surg Obes Relat Dis. 2007;3(6):597-601.
    1. Goldfine AB, Mun EC, Devine E, et al. . Patients with neuroglycopenia after gastric bypass surgery have exaggerated incretin and insulin secretory responses to a mixed meal. J Clin Endocrinol Metab. 2007;92(12):4678-4685.
    1. McLaughlin T, Peck M, Holst J, Deacon C. Reversible hyperinsulinemic hypoglycemia after gastric bypass: a consequence of altered nutrient delivery. J Clin Endocrinol Metab. 2010;95(4):1851-1855.
    1. Davis DB, Khoraki J, Ziemelis M, Sirinvaravong S, Han JY, Campos GM. Roux en Y gastric bypass hypoglycemia resolves with gastric feeding or reversal: Confirming a non-pancreatic etiology. Mol Metab. 2018;9:15-27.
    1. Kellogg TA, Bantle JP, Leslie DB, et al. . Postgastric bypass hyperinsulinemic hypoglycemia syndrome: characterization and response to a modified diet. Surg Obes Relat Dis. 2008;4(4):492-499.
    1. Suhl E, Anderson-Haynes SE, Mulla C, Patti ME. Medical nutrition therapy for post-bariatric hypoglycemia: practical insights. Surg Obes Relat Dis. 2017;13(5):888-896.
    1. Lee CJ, Brown T, Magnuson TH, Egan JM, Carlson O, Elahi D. Hormonal response to a mixed-meal challenge after reversal of gastric bypass for hypoglycemia. J Clin Endocrinol Metab. 2013;98(7):E1208-E1212.
    1. Vanderveen KA, Grant CS, Thompson GB, et al. . Outcomes and quality of life after partial pancreatectomy for noninsulinoma pancreatogenous hypoglycemia from diffuse islet cell disease. Surgery. 2010;148(6):1237-45; discussion 1245.
    1. Salehi M, Gastaldelli A, D’Alessio DA. Blockade of glucagon-like peptide 1 receptor corrects postprandial hypoglycemia after gastric bypass. Gastroenterology. 2014;146(3):669-680.e2.
    1. Craig CM, Liu LF, Deacon CF, Holst JJ, McLaughlin TL. Critical role for GLP-1 in symptomatic post-bariatric hypoglycaemia. Diabetologia. 2017;60(3):531-540.
    1. Craig CM, Liu LF, Nguyen T, Price C, Bingham J, McLaughlin TL. Efficacy and pharmacokinetics of subcutaneous exendin (9-39) in patients with post-bariatric hypoglycaemia. Diabetes Obes Metab. 2018;20(2):352-361.
    1. Tan M, Lamendola C, Luong R, McLaughlin T, Craig C. Safety, efficacy and pharmacokinetics of repeat subcutaneous dosing of avexitide (exendin 9-39) for treatment of post-bariatric hypoglycaemia. Diabetes Obes Metab. 2020;22(8):1406-1416.
    1. World Medical Association. World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA 2013;310( 20):2191-2194.
    1. International Hypoglycemia Study Group. Glucose concentrations of less than 3.0 mmol/L (54 mg/dL) should be reported in clinical trials: a joint position statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2017;40( 1):155-157.
    1. Danne T, Nimri R, Battelino T, et al. . International consensus on use of continuous glucose monitoring. Diabetes Care. 2017;40(12):1631-1640.
    1. Kjems LL, Holst JJ, Vølund A, Madsbad S. The influence of GLP-1 on glucose-stimulated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic subjects. Diabetes. 2003;52(2):380-386.
    1. Capozzi ME, Svendsen B, Encisco SE, et al. . β Cell tone is defined by proglucagon peptides through cAMP signaling. JCI Insight. 2019;4(5):e126742.
    1. Svendsen B, Larsen O, Gabe MBN, et al. . Insulin secretion depends on intra-islet glucagon signaling. Cell Rep. 2018;25(5):1127-1134.e2.
    1. Varma S, Clark JM, Schweitzer M, Magnuson T, Brown TT, Lee CJ. Weight regain in patients with symptoms of post-bariatric surgery hypoglycemia. Surg Obes Relat Dis. 2017;13(10):1728-1734.

Source: PubMed

3
Iratkozz fel