Skip to main content

Advertisement

Log in

Medical Nutrition Therapy and Other Approaches to Management of Post-bariatric Hypoglycemia: A Team-Based Approach

  • Metabolism (M Dalamaga, Section Editor)
  • Published:
Current Obesity Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

This manuscript provides a review of post-bariatric hypoglycemia (PBH) with a special focus on the role of the registered dietitian-nutritionist (RDN) and medical nutrition therapy (MNT) recommendations as foundational for management.

Recent Findings

As the number of bariatric surgeries rises yearly, with 256,000 performed in 2019, PBH is an increasingly encountered late complication. Following Roux-en-Y (RYGB) or vertical sleeve gastrectomy (VSG), about 1/3 of patients report symptoms suggestive of at least mild postprandial hypoglycemia, with severe and/or medically confirmed hypoglycemia in 1–10%. Anatomical alterations, changes in GLP1 and other intestinally derived hormones, excessive insulin response, reduced insulin clearance, impaired counterregulatory hormone response to hypoglycemia, and other factors contribute to PBH. MNT is the cornerstone of multidisciplinary treatment, with utilization of personal continuous glucose monitoring to improve safety when possible. While many individuals require pharmacotherapy, there are no currently approved medications for PBH.

Summary

Increasing awareness and identification of individuals at risk for or with PBH is critical given the potential impact on safety, nutrition, and quality of life. A team-based approach involving the individual, the RDN, and other clinicians is essential in providing ongoing assessment and individualization of MNT in the long-term management of PBH.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. American Society for Metabolic and Bariatric Surgery. Estimate of Bariatric Surgery Numbers, 2011–2019 | American Society for Metabolic and Bariatric Surgery. @asmbs. https://asmbs.org/resources/estimate-of-bariatric-surgery-numbers. Published 2021. Updated 2021–03–08. Accessed 3 Nov 2021.

  2. Mingrone G, Panunzi S, De Gaetano A, et al. Bariatric-metabolic surgery versus conventional medical treatment in obese patients with type 2 diabetes: 5 year follow-up of an open-label, single-centre, randomised controlled trial. Lancet (London, England). 2015;386(9997):964–73.

    Article  Google Scholar 

  3. Purnell JQ, Dewey EN, Laferrère B, et al. Diabetes remission status during seven-year follow-up of the longitudinal assessment of bariatric surgery study. J Clin Endocrinol Metab. 2021;106(3):774–88.

    Article  Google Scholar 

  4. Roth AE, Thornley CJ, Blackstone RP. Outcomes in bariatric and metabolic surgery: an updated 5-year review. Curr Obes Rep. 2020;9:380–9.

    Article  CAS  Google Scholar 

  5. Capristo E, Panunzi S, De Gaetano A, et al. Incidence of hypoglycemia after gastric bypass vs sleeve gastrectomy: a randomized trial. J Clin Endocrinol Metab. 2018;103(6):2136–46.

    Article  Google Scholar 

  6. Lazar LO, Sapojnikov S, Pines G, et al. Symptomatic and asymptomatic hypoglycemia post three different bariatric procedures: a common and severe complication. Endocr Pract. 2019. https://doi.org/10.4158/EP-2019-0185.

    Article  Google Scholar 

  7. Abrahamsson N, Edén Engström B, Sundbom M, Karlsson FA. Hypoglycemia in everyday life after gastric bypass and duodenal switch. Eur J Endocrinol. 2015;173(1).

  8. • Kubota T, Shoda K, Ushigome E, et al. Utility of continuous glucose monitoring following gastrectomy. Gastric Cancer. 2020;23(4):699–706. This study utilized CGM to show glycemic profiles of patients undergoing gastrectomy. CGM demonstrated nocturnal low sensor glucoses, as well as diurnal glycemic variability, not always associated with early dumping, thus implicating other mechanistic factors. Some participants were asymptomatic during periods of low sensor glucose.

    Article  CAS  Google Scholar 

  9. Zaloga GP, Chernow B. Postprandial hypoglycemia after Nissen fundoplication for reflux esophagitis. Gastroenterology. 1983;84(4):840–2.

    Article  CAS  Google Scholar 

  10. Bairdain S, Cleary M, Lien C, Vernon AH, Linden BC, Lautz DB. Laparoscopic adjustable gastric banding and hypoglycemia. Case Rep Endocrinol. 2013;2013:671848.

    Google Scholar 

  11. • Fischer LE, Wolfe BM, Fino N, et al. Postbariatric hypoglycemia: symptom patterns and associated risk factors in the Longitudinal Assessment of Bariatric Surgery study. Surg Obes Relat Dis. 2021;17(10):1787–98. This longitudinal cohort study examined hypoglycemia symptoms following RYGB and laparoscopic adjustable gastric banding. Preoperative symptoms consistent with hypoglycemia, and SSRI/SNRI use in those with no history of diabetes were associated with an increased risk of symptoms after RYGB.

    Article  Google Scholar 

  12. • Salehi M, Vella A, McLaughlin T, Patti ME. Hypoglycemia after gastric bypass surgery: current concepts and controversies. J Clin Endocrinol Metab. 2018;103(8):2815–26. This review summarizes the pathophysiology, diagnosis, and treatment of PBH.

    Article  Google Scholar 

  13. Nguyen NQ, Debreceni TL, Bambrick JE, et al. Rapid gastric and intestinal transit is a major determinant of changes in blood glucose, intestinal hormones, glucose absorption and postprandial symptoms after gastric bypass. Obesity (Silver Spring). 2014;22(9):2003–9.

    Article  CAS  Google Scholar 

  14. 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.e662.

    Article  CAS  Google Scholar 

  15. Dirksen C, Eiken A, Bojsen-Moller KN, et al. No islet cell hyperfunction, but altered gut-islet regulation and postprandial hypoglycemia in glucose-tolerant patients 3 years after gastric bypass surgery. Obes Surg. 2016;26(9):2263–7.

    Article  Google Scholar 

  16. Patti ME, Goldfine AB. Hypoglycemia after gastric bypass: the dark side of GLP-1. Gastroenterology. 2014;146(3):605–8.

    Article  CAS  Google Scholar 

  17. 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):2308–14.

    Article  CAS  Google Scholar 

  18. Zhou LY, Deng MQ, Xiao XH. Potential contribution of the gut microbiota to hypoglycemia after gastric bypass surgery. Chin Med J. 2020;133(15):1834–43.

    Article  Google Scholar 

  19. Mulla CM, Goldfine AB, Dreyfuss JM, et al. Plasma FGF-19 levels are increased in patients with post-bariatric hypoglycemia. Obes Surg. 2019;29(7):2092–9.

    Article  Google Scholar 

  20. Dror E, Dalmas E, Meier DT, et al. Postprandial macrophage-derived IL-1β stimulates insulin, and both synergistically promote glucose disposal and inflammation. Nat Immunol. 2017;18(3):283–92.

    Article  CAS  Google Scholar 

  21. Abrahamsson N, Börjesson JL, Sundbom M, Wiklund U, Karlsson FA, Eriksson JW. Gastric bypass reduces symptoms and hormonal responses in hypoglycemia. Diabetes. 2016;65(9):2667–75.

    Article  CAS  Google Scholar 

  22. •• Øhrstrøm CC, Hansen DL, Kielgast UL, et al. Counterregulatory responses to postprandial hypoglycemia after Roux-en-Y gastric bypass. Surg Obes Relat Dis. 2021;17(1):55–63. This study evaluated counterregulatory responses during postprandial hypoglycemia in individuals with PBH who underwent RYGB. Only minor increases in counterregulatory hormones were observed at the time of nadir glucose. The authors conclude that recurrent hypoglycemia may alter the glycemic threshold for counterregulatory activation.

    Article  Google Scholar 

  23. Martín-Timón I, del Cañizo-Gómez FJ. Mechanisms of hypoglycemia unawareness and implications in diabetic patients. World J Diabetes. 2015;6(7):912–26.

    Article  Google Scholar 

  24. Tack J, Arts J, Caenepeel P, De Wulf D, Bisschops R. Pathophysiology, diagnosis and management of postoperative dumping syndrome. Nat Rev Gastroenterol Hepatol. 2009;6(10):583–90.

    Article  Google Scholar 

  25. Marsk R, Jonas E, Rasmussen F, Näslund E. Nationwide cohort study of post-gastric bypass hypoglycaemia including 5,040 patients undergoing surgery for obesity in 1986–2006 in Sweden. Diabetologia. 2010;53(11):2307–11.

    Article  CAS  Google Scholar 

  26. Lee C, Brown T, Schweitzer M, Magnuson T, Clark J. 389-P: comparison of hormonal response to a mixed-meal challenge in individuals with hypoglycemia after sleeve gastrectomy vs. gastric bypass | Diabetes | American Diabetes Association. Diabetes. 2019;68.

  27. 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):492–9.

    Article  Google Scholar 

  28. Sarwar H, Chapman WH, Pender JR, et al. Hypoglycemia after Roux-en-Y gastric bypass: the BOLD experience. Obes Surg. 2014;24(7):1120–4.

    Article  Google Scholar 

  29. Lee CJ, Clark JM, Schweitzer M, Magnuson T, Steele K, Koerner O, Brown TT. Prevalence of and risk factors for hypoglycemic symptoms after gastric bypass and sleeve gastrectomy. Obesity (Silver Spring, Md). 2015;23(5):1079–84.

    Article  Google Scholar 

  30. 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):564–9.

    Article  Google Scholar 

  31. Whipple AO, Frantz VK. Adenoma of islet cells with hyperinsulinism: a review. Ann Surg. 1935;101(6):1299–335.

    Article  CAS  Google Scholar 

  32. Cryer PE, Axelrod L, Grossman AB, et al. Evaluation and management of adult hypoglycemic disorders: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2009;94(3):709–28.

    Article  CAS  Google Scholar 

  33. Lev-Ran A, Anderson RW. The diagnosis of postprandial hypoglycemia. Diabetes. 1981;30(12):996–9.

    Article  CAS  Google Scholar 

  34. Singh E, Vella A. Hypoglycemia after gastric bypass surgery. Diabetes Spectr. 2012;25(4):217–21.

    Article  CAS  Google Scholar 

  35. Andrade HF, Pedrosa W, Diniz Mde F, Passos VM. Adverse effects during the oral glucose tolerance test in post-bariatric surgery patients. Arch Endocrinol Metab. 2016;60(4):307–13.

    Article  Google Scholar 

  36. Ravindra S, Shetty S. Insulin autoimmune syndrome: a rare cause of spontaneous hypoglycaemia in non-diabetic individuals. BMJ case reports. 2020;13(12):e234448.

    Article  Google Scholar 

  37. Mulla CM, Storino A, Yee EU, et al. Insulinoma after bariatric surgery: diagnostic dilemma and therapeutic approaches. Obes Surg. 2016;26(4):874–81.

    Article  Google Scholar 

  38. Edwards K, de La Portilla L, Mirza FS, Luthra P. Proinsulin-predominant pancreatic neuroendocrine tumor-induced hypoglycemia after Roux-en-Y gastric bypass surgery. AACE Clin Case Rep. 2019;5(6):e339–43.

    Article  Google Scholar 

  39. Garla V, Sonani H, Palabindala V, Gomez-Sanchez C, Subauste J, Lien LF. Non-islet cell hypoglycemia: case series and review of the literature. Front Endocrinol. 2019;10:713–22.

    Article  Google Scholar 

  40. • van Furth AM, van den Broek M, Emous M, de Heide LJM, Kuipers F, van Beek AP. Cholecystectomy increases the risk of dumping syndrome and postbariatric hypoglycemia after bariatric surgery. Surg Obes Relat Dis. 2020;16(12):1939–47. This study compared the prevalence of self-reported complaints of DS and PBH in post-bariatric patients with and without cholecystectomy. The risk ratio for PBH was found to be higher after cholecystectomy post-sleeve gastrectomy, potentially related to altered bile acid kinetics.

    Article  Google Scholar 

  41. Lee CJ, Craig Wood G, Lazo M, et al. Risk of post-gastric bypass surgery hypoglycemia in nondiabetic individuals: a single center experience. Obesity (Silver Spring). 2016;24(6):1342–8.

    Article  CAS  Google Scholar 

  42. Rebelos E, Moriconi D, Scalese M, et al. Impact of postprandial hypoglycemia on weight loss after bariatric surgery. Obes Surg. 2020;30(6):2266–73.

    Article  Google Scholar 

  43. 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):0797–802.

    Article  Google Scholar 

  44. •• 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):888–96. This is a comprehensive review of nutritional strategies for PBH.

    Article  Google Scholar 

  45. Tricò D, Filice E, Trifirò S, Natali A. Manipulating the sequence of food ingestion improves glycemic control in type 2 diabetic patients under free-living conditions. Nutr Diabetes. 2016;6:e226.

    Article  Google Scholar 

  46. Mechanick JI, Apovian C, Brethauer S, et al. Clinical practice guidelines for the perioperative nutrition, metabolic, and nonsurgical support of patients undergoing bariatric procedures - 2019 update: cosponsored by American Association of Clinical Endocrinologists/American College of Endocrinology, the Obesity Society, American Society for Metabolic & Bariatric Surgery, Obesity Medicine Association, and American Society of Anesthesiologists - Executive Summary. Endocr Pract. 2019;25(12):1346–59.

    Google Scholar 

  47. Aguilera JM. The food matrix: implications in processing, nutrition and health. Crit Rev Food Sci Nutr. 2019;59(22):3612–29.

    Article  CAS  Google Scholar 

  48. Capuano E. The behavior of dietary fiber in the gastrointestinal tract determines its physiological effect. Crit Rev Food Sci Nutr. 2017;57(16):3543–64.

    Article  CAS  Google Scholar 

  49. •• Scarpellini E, Arts J, Karamanolis G, et al. International consensus on the diagnosis and management of dumping syndrome. Nat Rev Endocrinol. 2020;16(8):448–66. This is a consensus paper by a panel of international multidisciplinary experts which provides guidelines assessment/testing, diagnosis, and treatment of dumping syndrome.

    Article  Google Scholar 

  50. Field JB. Exercise and deficient carbohydrate storage and intake as causes of hypoglycemia. Endocrinol Metab Clin North Am. 1989;18(1):155–61.

    Article  CAS  Google Scholar 

  51. Gremse DA, Bucuvalas JC, Balistreri WF. Efficacy of cornstarch therapy in type III glycogen-storage disease. Am J Clin Nutr. 1990;52(4):671–4.

    Article  CAS  Google Scholar 

  52. •• Della Pepa G, Vetrani C, Lupoli R, et al. Uncooked cornstarch for the prevention of hypoglycemic events. Crit Rev Food Sci Nutr. 2021;18:1–14. This review summarizes the current evidence on the utility of uncooked cornstarch (UCS) in the treatment of hypoglycemia, including dose requirements, side effects, limitations, and plausible mechanisms by which UCS could prevent hypoglycemia.

    Google Scholar 

  53. Kaidar-Person O, Person B, Szomstein S, Rosenthal RJ. Nutritional deficiencies in morbidly obese patients: a new form of malnutrition? Part A: vitamins. Obes Surg. 2008;18(7):870–6.

    Article  Google Scholar 

  54. Roust LR, DiBaise JK. Nutrient deficiencies prior to bariatric surgery. Curr Opin Clin Nutr Metab Care. 2017;20(2):138–44.

    Article  CAS  Google Scholar 

  55. Kaidar-Person O, Person B, Szomstein S, Rosenthal RJ. Nutritional deficiencies in morbidly obese patients: a new form of malnutrition? Part B: minerals. Obes Surg. 2008;18(8):1028–34.

    Article  Google Scholar 

  56. •• Osland E, Powlesland H, Guthrie T, Lewis CA, Memon MA. Micronutrient management following bariatric surgery: the role of the dietitian in the postoperative period. Ann Transl Med. 2020;8(Suppl 1):S9. This review describes the anatomical, physiological, nutritional, and behavioral reasons for micronutrient vulnerabilities following bariatric surgery, and provides recommendations for routine monitoring and supplementation/replacement.

    Article  CAS  Google Scholar 

  57. • Wilson RB. Pathophysiology, prevention, and treatment of beriberi after gastric surgery. Nutr Rev. 2020;78(12):1015–29. This review discusses beriberi as an important nutritional deficiency after bariatric or gastric surgery, and provides recommendations for nutritional follow-up, assessment, and supplementation of thiamine.

    Article  Google Scholar 

  58. • Gasmi A, Bjørklund G, Mujawdiya PK, et al. Micronutrients deficiences in patients after bariatric surgery. Eur J Nutr. 2021;61(1):55–67. This systematic review identified literature related to multiple micronutrient and trace mineral deficiencies occurring after bariatric surgery. Post-surgical patients require additional doses of prophylactic supplementation chronically to maintain optimal micronutrient status; regular monitoring of serum nutrient levels starting at 3 months after surgery and periodically thereafter is recommended.

    Article  Google Scholar 

  59. Juhasz-Pocsine K, Rudnicki SA, Archer RL, Harik SI. Neurologic complications of gastric bypass surgery for morbid obesity. Neurology. 2007;68(21):1843–50.

    Article  Google Scholar 

  60. Briani C, Dalla Torre C, Citton V, et al. Cobalamin deficiency: clinical picture and radiological findings. Nutrients. 2013;5:4521–39.

    Article  Google Scholar 

  61. Lonsdale D. Dysautonomia, a heuristic approach to a revised model for etiology of disease. Evid Based Complement Alternat Med. 2009;6:3–10.

    Article  Google Scholar 

  62. Wadhwania R. Is vitamin D deficiency implicated in autonomic dysfunction? J Pediatr Neurosci. 2017;12:119–23.

    Article  Google Scholar 

  63. Meyer C, Grossmann R, Mitrakou A, et al. Effects of autonomic neuropathy on counterregulation and awareness of hypoglycemia in type 1 diabetic patients. Diabetes Care. 1998;21(11):1960–6.

    Article  CAS  Google Scholar 

  64. • Addison P, Carsky K, Patti ME, Roslin M. Hypoglycemia and dysautonomia after bariatric surgery: a systematic review and perspective. Obes Surg. 2022;32(5):1681–8. This systematic review discusses the diagnosis and treatment of dysautonomia after bariatric surgery.

    Article  Google Scholar 

  65. Lupoli R, Lembo E, Saldalamacchia G, Avola CK, Angrisani L, Capaldo B. Bariatric surgery and long-term nutritional issues. World J Diabetes. 2017;8(11):464–74.

    Article  Google Scholar 

  66. Cadegiani FA, Silva OS. Acarbose promotes remission of both early and late dumping syndromes in post-bariatric patients. Diabetes Metab Syndr Obes. 2016;9:443–6.

    Article  CAS  Google Scholar 

  67. Øhrstrøm CC, Worm D, Højager A, et al. Postprandial hypoglycaemia after Roux-en-Y gastric bypass and the effects of acarbose, sitagliptin, verapamil, liraglutide and pasireotide. Diabetes Obes Metab. 2019;21(9):2142–51.

    Article  Google Scholar 

  68. Spanakis E, Gragnoli C. Successful medical management of status post-Roux-en-Y-gastric-bypass hyperinsulinemic hypoglycemia. Obes Surg. 2009;19(9):1333–4.

    Article  Google Scholar 

  69. Gonzalez-Gonzalez A, Delgado M, Fraga-Fuentes MD. Use of diazoxide in management of severe postprandial hypoglycemia in patient after Roux-en-Y gastric bypass. Surg Obes Relat Dis. 2013;9(1):e18-19.

    Article  Google Scholar 

  70. Myint KS, Greenfield JR, Farooqi IS, Henning E, Holst JJ, Finer N. Prolonged successful therapy for hyperinsulinaemic hypoglycaemia after gastric bypass: the pathophysiological role of GLP1 and its response to a somatostatin analogue. Eur J Endocrinol. 2012;166(5):951–5.

    Article  CAS  Google Scholar 

  71. Mohammadi A, Sulaiman RA, Grossman AB. Pasireotide and octreotide in the treatment of severe late dumping syndrome. Clin Case Rep. 2017;5:1608–11.

    Article  Google Scholar 

  72. Xeris Pharmaceuticals I. Xeris Pharmaceuticals doses first patient in phase 2 trial evaluating its developmental ready-to-use glucagon in patients at risk from hypoglycemia following bariatric surgery. Xeris Pharmaceuticals, Inc. https://www.xerispharma.com/news/press-release/xeris-doses-first-patient-in-phase-2-trial. Published 2020. Accessed 16 Jun 2020.

  73. Knop FK. Treatment of post-bariatric hypoglycaemia (SHERRY). NIH, U.S. National Library of Medicine. https://clinicaltrials.gov/ct2/show/NCT04836273. Published 2021. Accessed 2 Nov 2021.

  74. Mulla CM, Zavitsanou S, Laguna Sanz AJ, et al. A randomized, placebo-controlled double-blind trial of a closed-loop glucagon system for postbariatric hypoglycemia. J Clin Endocrinol Metab. 2020;105(4):e1260–71.

    Article  Google Scholar 

  75. Martinussen C, Veedfald S, Dirksen C, et al. The effect of acute dual SGLT1/SGLT2 inhibition on incretin release and glucose metabolism after gastric bypass surgery. Am J Physiol Endocrinol Metab. 2020;318(6):e956–64.

    Article  CAS  Google Scholar 

  76. •• Craig CM, Lawler HM, Lee CE, et al. PREVENT: a randomized, placebo-controlled crossover trial of avexitide for treatment of postbariatric hypoglycemia. J Clin Endocrinol Metab. 2021;106(8):e3235–48. Avexitide was found to be well tolerated and resulted in robust and consistent improvements across several metabolic and clinical parameters, as assessed during MMTT, self-monitoring of capillary glucose, and CGM.

    Article  Google Scholar 

  77. Abrahamsson N, Engström BE, Sundbom M, Karlsson FA. GLP1 analogs as treatment of postprandial hypoglycemia following gastric bypass surgery: a potential new indication? Eur J Endocrinol. 2013;169(6):885–9.

    Article  CAS  Google Scholar 

  78. 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.

    Article  Google Scholar 

  79. Ames A, Lago-Hernandez CA, Grunvald E. Hypoglycemia after gastric bypass successfully treated with calcium channel blockers: two case reports. J Endocr Soc. 2019;3:1417–22.

    Article  Google Scholar 

  80. 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.

    Article  CAS  Google Scholar 

  81. 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):1851–5.

    Article  CAS  Google Scholar 

  82. Craig CM, Lamendola C, Holst JJ, Deacon CF, McLaughlin TL. The use of gastrostomy tube for the long-term remission of hyperinsulinemic hypoglycemia after Roux-en-Y gastric bypass: a case report. AACE Clin Case Rep. 2015;1(2):e84–7.

    Article  Google Scholar 

  83. Arora I, Patti ME. Can reversal of RYGB also reverse hypoglycemia? Mol Metab. 2018;9:1–3.

    Article  Google Scholar 

  84. Qvigstad E, Gulseth HL, Risstad H, et al. A novel technique of Roux-en-Y gastric bypass reversal for postprandial hyperinsulinemic hypoglycaemia: a case report. Int J Surg Case Rep. 2016;21:91–4.

    Article  CAS  Google Scholar 

  85. 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):E1208-1212.

    Article  CAS  Google Scholar 

  86. Ma P, Ghiassi S, Lloyd A, et al. Reversal of Roux en Y gastric bypass: largest single institution experience. Surg Obes Relat Dis. 2019;15(8):1311–6.

    Article  Google Scholar 

  87. Shoar S, Nguyen T, Ona MA, et al. Roux-en-Y gastric bypass reversal: a systematic review. Surg Obes Relat Dis. 2016;12(7):1366–72.

    Article  Google Scholar 

  88. Ahmed AA. Hypoglycemia and safe driving. Ann Saudi Med. 2010;30(6):464–7.

    Article  Google Scholar 

Download references

Funding

MEP gratefully acknowledges grant support related to this project from National Institutes of Health R01 DK121995 and P30 DK036836 (DRC, Joslin Diabetes Center).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Nicole Patience or Mary Elizabeth Patti.

Ethics declarations

Conflict of Interest

NP, AS, and CC declare that they have no conflict of interest related to this manuscript. MEP reports personal fees from Astra Zeneca, Fractyl, Hanmi Pharmaceutical, MBX Biosciences, Recordati, Poxel, Eiger Pharmaceuticals, and Xeris, and grants from Chan-Zuckerberg Initiative, Dexcom, and Helmsley Trust, outside the submitted work.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Metabolism

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (PPTX 54 kb)

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Patience, N., Sheehan, A., Cummings, C. et al. Medical Nutrition Therapy and Other Approaches to Management of Post-bariatric Hypoglycemia: A Team-Based Approach. Curr Obes Rep 11, 277–286 (2022). https://doi.org/10.1007/s13679-022-00482-0

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13679-022-00482-0

Keywords

Navigation