GLP-1 analogue liraglutide as adjunct treatment in diabetes type 2 after failed bariatric/metabolic surgery
Editorial Commentary

GLP-1 analogue liraglutide as adjunct treatment in diabetes type 2 after failed bariatric/metabolic surgery

Per M. Hellström

Department of Medical Sciences, Uppsala University, Uppsala, Sweden

Correspondence to: Prof. Per M. Hellström. Department of Medical Sciences, Uppsala University Hospital, Bldg 40, 5th floor, SE-75185 UPPSALA, Sweden. Email: Per.Hellstrom@medsci.uu.se.

Provenance: This is an invited article commissioned by the Academic Editor Jiewen Jin (Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China).

Comment on: Miras AD, Pérez-Pevida B, Aldhwayan M, et al. Adjunctive liraglutide treatment in patients with persistent or recurrent type 2 diabetes after metabolic surgery (GRAVITAS): a randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol 2019;7:549-59.


Submitted Aug 10, 2019. Accepted for publication Aug 22, 2019.

doi: 10.21037/atm.2019.08.94


Obesity with its negative effect on health (1) has become a major challenge to physicians as well as surgeons throughout the world. Today more than 650 million people worldwide are obese (1). To the physician, the individuals’ positive energy balance with a greater energy intake than expenditure means an increased rate of diabetes type 2 and insulin resistance along with debilitating complications such as hypertension, stroke and cardiac disease, sleep respiratory disorders including sleep disturbance, blood lipid abnormalities and non-alcoholic fatty liver disease. To the surgeon, the problem mainly deals with the stigmatizing body composition, excessive accumulation of adipose tissue and immobility. Furthermore, there is a link to a number of cancers, such as endometrium, breast, ovary, prostate, liver, gallbladder, kidney, and colorectal cancer.

Obesity is most commonly caused by a combination of over-eating, limited physical activity, and genetic susceptibility (1,2). Metabolically, obese people have a greater energy expenditure than their normal counterparts due to the huge energy required to maintain an increased body mass in a vicious circle (3,4). The excessive food intake will put an extra requirement on the capacity of inner organs, such as the liver and pancreas, in order to meet the unrestricted load of nutrients that is delivered through the stomach and small intestine. Therefore, food restriction seems to be the obvious way to tackle the problem and move forward. At the individual level, WHO recommends limitation of the energy intake with less total fats and sugars as well as increased intake of fruit, vegetables and legumes, as well as whole grains and nuts (1). Experience is telling that this is hard to come by.

In the clinic, bariatric surgery is today the only method considered effective and reliable enough to achieve substantial weight loss and relief of obesity-related co-morbidities, with superior outcomes as compared to dieting (5,6). Gastric bypass surgery (7) usually results in durable weight loss (8), along with marked hormonal changes that develop immediately after surgery, even before demonstrable weight loss takes place (9). As an alternative, sleeve gastrectomy as a stand-alone procedure has gained popularity to achieve large weight loss (10). Sleeve gastrectomy is considered as effective as gastric bypass surgery, including improvements in glucose homeostasis even before substantial weight loss ensues. The two operations are both permanent, reduce hunger and cause similar weight loss. The weight-loss independent benefits are related to a diminished gastric volume, gut peptide release, and expression of genes entangled in intestinal glucose absorption (11,12).

Through research on gastrointestinal peptide hormones, accumulating evidence show that there is also a pharmacological way forward to bring about weight loss. Glucagon-like peptide-1 (GLP-1) is an anorexigenic and insulinogenic hormone and neuropeptide released from the distal gastrointestinal tract. GLP-1 slows gastric emptying, an effect that may be due hormonal as well as transmitter actions within the myenteric plexus (13,14). Postprandial GLP-1 levels are suppressed in obese subjects as compared to the lean (9). Fasting levels of GLP-1 increase after bypass surgery as well as after diet-induced weight loss, and stay high after even one year of weight maintenance (9,15). Studies employing GLP-1 analogues have shown liraglutide to be effective for weight-reducing purposes, even though a relatively high dose of 3 mg was used to achieve a significant weight loss of 8% after 56 weeks (16). Recently, data was launched showing that another analogue, semaglutide, at a daily dose of 0.4 mg was able to further reduce body weight by 16.2% (17). This pharmacological development makes GLP-1 analogues really interesting for weight reduction purposes and true competitors to surgery. To this end, since the metabolic effects of bariatric surgery aside from weight control are strong, the term metabolic surgery has now been coined for the enhanced metabolic control and glucose homeostasis after bariatric surgery.

In The Lancet Diabetes & Endocrinology, Alexander D. Miras and colleagues report the results of a well-conducted randomised, double-blind, placebo-controlled clinical trial on the adjunctive use of liraglutide as a treatment in patients with persistent or recurrent type 2 diabetes after metabolic surgery (18). The basic standpoint for the article is the fact that after surgery 30–63% of diabetic patients attain remission to normoglycaemia without any additional glucose-lowering medications, but also that 37–70% do not attain remission. Furthermore, in those who achieve normoglycaemia to remission, 21–43% relapse within 3–9 years (19,20). Hence, the management of persistent or recurrent diabetes type 2 after metabolic surgery represents a clinical challenge because post-surgical conditions may compromise regular pharmacokinetics with erratic drug absorption, why an injectable such as liraglutide would be the natural choice. However, in the absence of a randomized trial this condition still leaves the diabetologist with no available guidelines or specific recommendations for further treatment. Against this background the GRAVITAS (GLP-1 Receptor Agonist interVentIon for poor responders afTer bariAtric Surgery) clinical trial was set out to examine the safety and efficacy of subcutaneous injections of the GLP-1 receptor agonist liraglutide on glycaemic control in patients with persistent or recurrent diabetes type 2 after bariatric Roux-en-Y gastric bypass or vertical sleeve gastrectomy.

All in all, 80 adult participants out of a total number of 303 pre-screened individuals, were recruited who had undergone bariatric surgery at least 12 months before enrolment, had diabetes type 2 with HbA1c over 48 mmol/mol (>6.5%), treatment with metformin or long-acting insulin, and had given informed consent. Of those assigned, 71 completed the trial reaching a statistical power of at least 80% at the P<0.05 level. The study design was parallel with an initial run-in period for assessing the patients’ adherence to self-administration of the injectable study drug or placebo once daily. Then, the participants were randomised to either liraglutide or placebo in a 2:1 fashion, starting with a daily dose of 0.6 mg liraglutide increasing 0.6 mg per week up to 1.8 mg or maximum tolerated dose for 26 weeks in a row. All patients were overseen by a diabetologist-endocrinologist, dietician, and psychiatrist. The key aspect of the simultaneous nutritional approach aimed at an intake deficit of 500 kcal per day from their baseline consumption, with advice on adequate intake of protein, vegetables and fruit, along with reduced amounts of high-glycaemic index foods, alcohol, salt and saturated fat.

The primary outcome of the study was the change in HbA1c after 26 weeks of treatment. The secondary outcomes were assessment of changes from baseline in bodyweight, systolic and diastolic blood pressure, blood lipid profile, the number of glucose-lowering drugs, the number of patients on insulin and insulin dosage, the obesity-related co-morbidity score, behavioural traits, quality of life, sweet taste detection, food reward ratings, the number of hypoglycaemic episodes, and collected adverse events assessed by reporting at each trial study visit.

There were no differences in baseline features or properties between patients in the liraglutide and placebo groups. Over the treatment period there was a parallel decrease in HbA1c and body weight throughout the study reaching highly significant nadirs at 18 and 26 weeks, respectively. The effect on HbA1c with a mean difference of –13.3 mmol/mol was striking as was the mean difference of in body weight between the two groups with –5.26 kg. Other outcomes did not show any significant changes.

The strength of this study is the generalizability that is shown by the similar response rate to liraglutide irrespective of the type of metabolic surgery. Since the two operations, i.e., gastric bypass and sleeve gastrectomy, are so divergent in nature, with one restricting the stomach size to a minimum, and the other leaving a considerable part of the stomach in place, it seems that a common mechanism of action of liraglutide (and similar GLP-1 analogues) on gastric functions is unlikely. The fact that both operations would permit a rapid delivery of foodstuffs into the small intestine where a host of gastrointestinal peptides [e.g., incretins such as glucose-dependent insulinotropic peptide (GIP) and GLP-1] are released, opens the possibility of a common peripheral mechanism of action. Such a mechanism would be expected to cause dumping effects but no adverse effects alike were found in the study. The paucity of a dumping syndrome rules out this possibility as a weight-reducing mechanism. Hence, it would have been of interest to see how the bioavailability of various nutrients or pharmaceuticals behave under these two circumstances. Experiments using glucose or paracetamol for absorption tests would possibly give some more insight into the mechanism of action of liraglutide. Even if peripheral mechanisms may be in operation for weight control, a centrally-mediated action of liraglutide, primarily in the nucleus of the solitary tract, may also be necessary in order to accomplish the substantial weight loss seen with different GLP-1 analogues. To summarize, irrespective of the type of previous metabolic surgery, treatment with liraglutide at doses of 1.2–1.8 mg per day seems to overcome persistent or recurrent diabetes type 2 after bariatric surgery along with a needful weight loss.

In terms of weight loss, bariatric surgery is no guarantee for success. Experience show that even with highly standardized surgery which results in a maintained weight loss for most patients (8) some degree of postsurgical weight regain is commonplace, as noted in one-half of patients two years after surgery (21). Results show that after 5 years, about one-fifth of operated patients will have lost no more than half of their preoperative overweight (22), considered a hallmark of failing bariatric surgery (23). Amongst gut peptide hormones in the proximal gastrointestinal tract, ghrelin is higher in the obese than in normal-weight individuals (24), whereas GLP-1 is low (9,14). In a recent study of non-responders to bariatric surgery, an early rise of ghrelin to baseline levels in conjunction with decreasing leptin levels below baseline during an oral glucose tolerance test have been suggested as a possible appetite-stimulating effect, thus contributing to undesired weight gain after surgery (25).

The present study by Miras and co-workers shows that the GLP-1 mechanism as brought about by liraglutide is able to overcome basic pathophysiological aberrations in obese subjects with low plasma GLP-1 levels (9,14). The stimulation of GLP-1 receptors controls not only glucose homeostasis, but also body weight maintenance with a very limited risk of adverse effects (18). Even if change in bodyweight was stated as a secondary outcome of the study, anyone skilled in the field would recognise this as an important coeffect to the reduced HbA1c. However, in order to make this finding a fully acceptable discovery, a clinical study according to the regulatory construct would require the change in body weight as a primary study endpoint. There is no single superior method of how to evaluate postoperative weight results. Measurements of change in excess BMI, total body weight and degree of weight regain are all different approaches developed for this purpose. The percent of excess BMI loss (%EBMIL), although criticized for giving better results in lightweight subjects as compared to the heavyweights, is today considered a reliable measurement as primary endpoint of weight regain after weight loss. In addition, beyond changes in %EBMIL, successful weight loss should also comprise remission of co-morbidities and improvement of quality of life as secondary endpoints. Since we today much too often meet patients who experience poor long-term weight results after bariatric surgery, and since we do not have a clear picture of the underlying mechanisms for surgery failure, treatment with GLP-1 analogues, be it liraglutide or semaglutide, or similar compounds, stand out as effective treatment with little side-effect burden, largely limited to nausea and constipation, which might be easily overcome by dose-adjustments in the clinical setting.


Acknowledgments

None.


Footnote

Conflicts of Interest: The author has no conflicts of interest to declare.

Ethical Statement: The author is accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.


References

  1. Obesity and overweight fact sheet N°311. WHO. February 2018. Retrieved August 10, 2019. Available online: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight
  2. Yazdi FT, Clee SM, Meyre D. Obesity genetics in mouse and human: back and forth, and back again. PeerJ 2015;3:e856. [Crossref] [PubMed]
  3. Kushner R. Treatment of the obese patient. In: Contemporary Endocrinology. Totowa, NJ: Humana Press, 2007:158.
  4. Oxford Handbook of Medical Sciences (2nd ed.). Oxford: OUP Oxford, 2011:180.
  5. Gloy VL, Briel M, Bhatt DL, et al. Bariatric surgery versus non-surgical treatment for obesity: a systematic review and meta-analysis of randomised controlled trials. BMJ (Clinical research ed) 2013;347:f5934. [Crossref] [PubMed]
  6. Colquitt JL, Pickett K, Loveman E, et al. Surgery for weight loss in adults. Cochrane Database Syst Rev 2014.CD003641. [PubMed]
  7. Angrisani L, Santonicola A, Iovino P, et al. Bariatric surgery worldwide 2013. Obes Surg 2015;25:1822-32. [Crossref] [PubMed]
  8. Maciejewski ML, Arterburn DE, Van Scoyoc L, et al. Bariatric surgery and long-term durability of weight loss. JAMA Surg 2016;151:1046-55. [Crossref] [PubMed]
  9. Falkén Y, Hellström PM, Holst JJ, et al. Changes in glucose homeostasis after Roux-en-Y gastric bypass surgery for obesity at day three, two months, and one year after surgery: role of gut peptides. J Clin Endocrinol Metab 2011;96:2227-35. [Crossref] [PubMed]
  10. Spiegel HU, Skawran S. From longitudinal gastric resection to sleeve gastrectomy--revival of a previously established surgical procedure. J Gastrointest Surg 2011;15:219-28. [Crossref] [PubMed]
  11. Hutch CR, Sandoval D. The role of GLP-1 in the metabolic success of bariatric surgery. Endocrinology 2017;158:4139-51. [Crossref] [PubMed]
  12. Cavin JB, Couvelard A, Lebtahi R, et al. Differences in alimentary glucose absorption and intestinal disposal of blood glucose after Roux-en-Y gastric bypass vs sleeve gastrectomy. Gastroenterology 2016;150:454-64.e9. [Crossref] [PubMed]
  13. Näslund E, Bogefors J, Skogar S, et al. GLP-1 slows solid gastric emptying and inhibits insulin, glucagon, and PYY release in humans. Am J Physiol 1999;277:R910-6. [PubMed]
  14. Adam TCM, Westerterp-Plantenga MS. Glucagon-like peptide-1 release and satiety after a nutrient challenge in normal-weight and obese subjects. Br J Nutr 2005;93:845-51. [Crossref] [PubMed]
  15. Iepsen EW, Lundgren J, Holst JJ, et al. Successful weight loss maintenance includes long-term increased meal responses of GLP-1 and PYY3-36. Eur J Endocrinol 2016;174:775-84. [Crossref] [PubMed]
  16. Pi-Sunyer X, Astrup A, Fujioka K, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N Engl J Med 2015;373:11-22. [Crossref] [PubMed]
  17. O'Neil PM, Birkenfeld AL, McGowan B, et al. Efficacy and safety of semaglutide compared with liraglutide and placebo for weight loss in patients with obesity: a randomised, double-blind, placebo and active controlled, dose-ranging, phase 2 trial. Lancet 2018;392:637-49. [Crossref] [PubMed]
  18. Miras AD, Pérez-Pevida B, Aldhwayan M, et al. Adjunctive liraglutide treatment in patients with persistent or recurrent type 2 diabetes after metabolic surgery (GRAVITAS): a randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol 2019;7:549-59. [Crossref] [PubMed]
  19. Rubino F, Nathan DM, Eckel RH, et al. Metabolic surgery in the treatment algorithm for type 2 diabetes: a joint statement by international diabetes organizations. Diabetes Care 2016;39:861-77. [Crossref] [PubMed]
  20. Yu J, Zhou X, Li L, et al. The long-term effects of bariatric surgery for type 2 diabetes: systematic review and meta-analysis of randomized and non-randomized evidence. Obes Surg 2015;25:143-58. [Crossref] [PubMed]
  21. Magro DO, Geloneze B, Delfini R, et al. Long-term weight regain after gastric bypass: A 5-year prospective study. Obes Surg 2008;18:648-51. [Crossref] [PubMed]
  22. SOReg. Year report 2015 - Part 2. Scandinavian Obesity Surgery Registry (SOReg) Available online: https://www.ucr.uu.se/soreg/component/edocman/2016-10-19-arsrapport-2015-del-2. Retrieved August 10, 2019.
  23. Reinhold RB. Critical analysis of long term weight loss following gastric bypass. Surg Gynecol Obstet 1982;155:385-94. [PubMed]
  24. Klok MD, Jakobsdottir S, Drent ML. The role of leptin and ghrelin in the regulation of food intake and body weight in humans: a review. Obes Rev 2007;8:21-34. [Crossref] [PubMed]
  25. Sima E, Webb DL, Hellström PM, Sundbom M. Non-responders After Gastric Bypass Surgery for Morbid Obesity: Peptide Hormones and Glucose Homeostasis. Obes Surg 2019. [Epub ahead of print]. [Crossref] [PubMed]
Cite this article as: Hellström PM. GLP-1 analogue liraglutide as adjunct treatment in diabetes type 2 after failed bariatric/metabolic surgery. Ann Transl Med 2019;7(Suppl 6):S240. doi: 10.21037/atm.2019.08.94

Download Citation