Skip to main content
Log in

The Future of Incretin-Based Approaches for Neurodegenerative Diseases in Older Adults: Which to Choose? A Review of their Potential Efficacy and Suitability

  • Leading Article
  • Published:
Drugs & Aging Aims and scope Submit manuscript

Abstract

The current treatment options for neurodegenerative diseases in older adults rely mainly on providing symptomatic relief. Yet, it remains imperative to identify agents that slow or halt disease progression to avoid the most disabling features often associated with advanced disease stages. A potential overlap between the pathological processes involved in diabetes and neurodegeneration has been established, raising the question of whether incretin-based therapies for diabetes may also be useful in treating neurodegenerative diseases in older adults. Here, we review the different agents that belong to this class of drugs (GLP-1 receptor agonists, dual/triple receptor agonists, DPP-4 inhibitors) and describe the data supporting their potential role in treating neurodegenerative conditions including Parkinson’s disease and Alzheimer’s disease. We further discuss whether there are any distinctive properties among them, particularly in the context of safety or tolerability and CNS penetration, that might facilitate their successful repurposing as disease-modifying drugs. Proof-of-efficacy data will obviously be of the greatest importance, and this is most likely to be demonstrable in agents that reach the central nervous system and impact on neuronal GLP-1 receptors. Additionally, however, the long-term safety and tolerability (including gastrointestinal side effects and unwanted weight loss) as well as the route of administration of this class of agents may also ultimately determine success and these aspects should be considered in prioritising which approaches to subject to formal clinical trial evaluations.

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.

Similar content being viewed by others

References

  1. Ahmed RM, Devenney EM, Irish M, Ittner A, Naismith S, Ittner LM, et al. Neuronal network disintegration: common pathways linking neurodegenerative diseases. J Neurol Neurosurg Psychiatry. 2016;87:1234–41.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Ott A, Stolk RP, van Harskamp F, Pols HAP, Hofman A, Breteler MMB. Diabetes mellitus and the risk of dementia: the Rotterdam Study. Neurology. 1999;53:1937–1937.

    Article  CAS  Google Scholar 

  3. Sandyk R. The relationship between diabetes mellitus and Parkinson’s disease. Int J Neurosci. 1993;69:125–30.

    Article  CAS  PubMed  Google Scholar 

  4. Xu W, Caracciolo B, Wang H-X, Winblad B, Backman L, Qiu C, et al. Accelerated progression from mild cognitive impairment to dementia in people with diabetes. Diabetes. 2010;59:2928–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Mukherjee A, Morales-Scheihing D, Butler PC, Soto C. Type 2 diabetes as a protein misfolding disease. Trends Mol Med. 2015;21:439–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kim J, Wei Y, Sowers JR. Role of mitochondrial dysfunction in insulin resistance. Circ Res. 2008;102:401–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Talbot K, Wang H-Y, Kazi H, Han L-Y, Bakshi KP, Stucky A, et al. Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J Clin Investig. 2012;122:1316–38.

    Article  CAS  PubMed  Google Scholar 

  8. Foltynie T, Athauda D. Repurposing anti-diabetic drugs for the treatment of Parkinson’s disease: rationale and clinical experience. 1st ed. Amsterdam: Elsevier; 2020.

    Google Scholar 

  9. Perry TA, Greig NH. The glucagon-like peptides: a new genre in therapeutic targets for intervention in Alzheimer’s disease. J Alzheimer’s Dis. 2002.

  10. Perry T, Haughey NJ, Mattson MP, Egan JM, Greig NH. Protection and reversal of excitotoxic neuronal damage by glucagon-like peptide-1 and exendin-4. J Pharmacol Exp Ther. 2002;302:881–8.

    Article  CAS  PubMed  Google Scholar 

  11. Perry T, Lahiri DK, Chen D, Zhou J, Shaw KTY, Egan JM, et al. A novel neurotrophic property of glucagon-like peptide 1: a promoter of nerve growth factor-mediated differentiation in PC12 cells. J Pharmacol Exp Ther. 2002;300:958–66.

    Article  CAS  PubMed  Google Scholar 

  12. Combs CK. Are GLP-1 receptor agonists useful against traumatic brain injury? J Neurochem. 2015.

  13. Maskery MP, Holscher C, Jones SP, Price ci, Strain WD, Watkins CL, et al. Glucagon-like peptide-1 receptor agonists as neuroprotective agents for ischemic stroke: a systematic scoping review. J Cereb Blood Flow Metab. 2021;41:14.

    Article  CAS  PubMed  Google Scholar 

  14. Martin B, Golden E, Carlson OD, Pistell P, Zhou J, Kim W, et al. Exendin-4 improves glycemic control, ameliorates brain and pancreatic pathologies, and extends survival in a mouse Model of Huntington’s disease. Diabetes. 2009.

  15. Li Y, Chigurupati S, Holloway HW, Mughal M, Tweedie D, Bruestle DA, et al. Exendin-4 ameliorates motor neuron degeneration in cellular and animal models of amyotrophic lateral sclerosis. PLoS One. 2012.

  16. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007;132:2131–57.

    Article  CAS  PubMed  Google Scholar 

  17. Gentilella R, Pechtner V, Corcos A, Consoli A. Glucagon-like peptide-1 receptor agonists in type 2 diabetes treatment: are they all the same? Diabetes Metab Res Rev. 2019;35:1–21.

    Article  CAS  Google Scholar 

  18. Egan JM, Bulotta A, Hui H, Perfetti R. GLP-1 receptor agonists are growth and differentiation factors for pancreatic islet beta cells. Diabetes Metab Res Rev. 2003;19:115–23.

    Article  CAS  PubMed  Google Scholar 

  19. Ekkelund Petersen K, Rakipovski G, Raun K, Lykkesfeldt J. Does glucagon-like peptide-1 ameliorate oxidative stress in diabetes? Evidence based on experimental and clinical studies. Curr Diabetes Rev. 2016;12:331–58.

    Article  PubMed  Google Scholar 

  20. Fan R, Li X, Gu X, Chan JCN, Xu G. Exendin-4 protects pancreatic beta cells from human islet amyloid polypeptide-induced cell damage: potential involvement of AKT and mitochondria biogenesis. Diabetes Obes Metab. 2010;12:815–24.

    Article  CAS  PubMed  Google Scholar 

  21. Buse JB, Henry RR, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in sulfonylurea-treated patients with type 2 diabetes. Diabetes Care. 2004;27:2628–35.

    Article  CAS  PubMed  Google Scholar 

  22. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care. 2005;28:1092–100.

    Article  CAS  PubMed  Google Scholar 

  23. Kendall DM, Riddle MC, Rosenstock J, Zhuang D, Kim DD, Fineman MS, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care. 2005;28:1083–91.

    Article  CAS  PubMed  Google Scholar 

  24. Rosenstock J, Raccah D, Koranyi L, Maffei L, Boka G, Miossec P, et al. Efficacy and safety of lixisenatide once daily versus exenatide twice daily in type 2 diabetes inadequately controlled on metformin: a 24-week, randomized, open-label, active-controlled study (GetGoal-X). Diabetes Care. 2013;36:2945–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Linnebjerg H, Park S, Kothare PA, Trautmann ME, Mace K, Fineman M, et al. Effect of exenatide on gastric emptying and relationship to postprandial glycemia in type 2 diabetes. Regul Pept. 2008;151:123–9.

    Article  CAS  PubMed  Google Scholar 

  26. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin. 2008;24:2943–52.

    Article  CAS  PubMed  Google Scholar 

  27. Drucker DJ, Buse JB, Taylor K, Kendall DM, Trautmann M, Zhuang D, et al. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet. 2008;372:1240–50.

    Article  CAS  PubMed  Google Scholar 

  28. Blevins T, Pullman J, Malloy J, Yan P, Taylor K, Schulteis C, et al. DURATION-5: Exenatide once weekly resulted in greater improvements in glycemic control compared with exenatide twice daily in patients with type 2 diabetes. J Clin Endocrinol Metab. 2011;96:1301–10.

    Article  CAS  PubMed  Google Scholar 

  29. Buse JB, Rosenstock J, Sesti G, Schmidt WE, Montanya E, Brett JH, et al. Liraglutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6). Lancet. 2009;374:39–47.

    Article  CAS  PubMed  Google Scholar 

  30. Buse JB, Nauck M, Forst T, Sheu WHH, Shenouda SK, Heilmann CR, et al. Exenatide once weekly versus liraglutide once daily in patients with type 2 diabetes (DURATION-6): a randomised, open-label study. Lancet. 2013;381:117–24.

    Article  CAS  PubMed  Google Scholar 

  31. Pratley RE, Nauck MA, Barnett AH, Feinglos MN, Ovalle F, Harman-Boehm I, et al. Once-weekly albiglutide versus once-daily liraglutide in patients with type 2 diabetes inadequately controlled on oral drugs (HARMONY 7): a randomised, open-label, multicentre, non-inferiority phase 3 study. Lancet Diabetes Endocrinol. 2014;2:289–97.

    Article  CAS  PubMed  Google Scholar 

  32. Dungan KM, Povedano ST, Forst T, González JGG, Atisso C, Sealls W, et al. Once-weekly dulaglutide versus once-daily liraglutide in metformin-treated patients with type 2 diabetes (AWARD-6): a randomised, open-label, phase 3, non-inferiority trial. Lancet. 2014;384:1349–57.

    Article  CAS  PubMed  Google Scholar 

  33. Wysham C, Blevins T, Arakaki R, Colon G, Garcia P, Atisso C, et al. Efficacy and safety of dulaglutide added onto pioglitazone and metformin versus exenatide in type 2 diabetes in a randomized controlled trial (AWARD-1). Diabetes Care. 2014;37:2159–67.

    Article  CAS  PubMed  Google Scholar 

  34. Aroda VR, Rosenstock J, Terauchi Y, Altuntas Y, Lalic NM, Morales Villegas EC, et al. PIONEER 1: Randomized clinical trial of the efficacy and safety of oral semaglutide monotherapy in comparison with placebo in patients with type 2 diabetes. Diabetes Care. 2019;42:1724–32.

    Article  CAS  PubMed  Google Scholar 

  35. Pratley R, Amod A, Hoff ST, Kadowaki T, Lingvay I, Nauck M, et al. Oral semaglutide versus subcutaneous liraglutide and placebo in type 2 diabetes (PIONEER 4): a randomised, double-blind, phase 3a trial. Lancet. 2019;394:39–50.

    Article  CAS  PubMed  Google Scholar 

  36. Husain M, Birkenfeld AL, Donsmark M, Dungan K, Eliaschewitz FG, Franco DR, et al. Oral semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2019;381:841–51.

    Article  CAS  PubMed  Google Scholar 

  37. Nauck MA, Bartels E, Orskov C, Ebert R, Creutzfeldt W. Additive insulinotropic effects of exogenous synthetic human gastric inhibitory polypeptide and glucagon-like peptide-1-(7–36) amide infused at near-physiological insulinotropic hormone and glucose concentrations. J Clin Endocrinol Metab. 1993;76:912–7.

    CAS  PubMed  Google Scholar 

  38. Bastin M, Andreelli F. Dual GIP–GLP1-receptor agonists in the treatment of type 2 diabetes: a short review on emerging data and therapeutic potential. Diabetes Metab Syndr Obes Targets Ther. 2019;12:1973–85.

    Article  CAS  Google Scholar 

  39. Schmitt C, Portron A, Jadidi S, Sarkar N, DiMarchi R. Pharmacodynamics, pharmacokinetics and safety of multiple ascending doses of the novel dual glucose-dependent insulinotropic polypeptide/glucagon-like peptide-1 agonist RG7697 in people with type 2 diabetes mellitus. Diabetes Obes Metab. 2017;19:1436–45.

    Article  CAS  PubMed  Google Scholar 

  40. Frias JP, Bastyr EJ, Vignati L, Tschöp MH, Schmitt C, Owen K, et al. The sustained effects of a dual GIP/GLP-1 receptor agonist, NNC0090-2746, in patients with type 2 diabetes. Cell Metab. 2017;26:343–52.

    Article  CAS  PubMed  Google Scholar 

  41. Coskun T, Sloop KW, Loghin C, Alsina-Fernandez J, Urva S, Bokvist KB, et al. LY3298176, a novel dual GIP and GLP-1 receptor agonist for the treatment of type 2 diabetes mellitus: from discovery to clinical proof of concept. Mol Metab. 2018;18:3–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Frias JP, Nauck MA, Van J, Kutner ME, Cui X, Benson C, et al. Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: a randomised, placebo-controlled and active comparator-controlled phase 2 trial. Lancet. 2018;392:2180–93.

    Article  CAS  PubMed  Google Scholar 

  43. Capozzi ME, DiMarchi RD, Tschöp MH, Finan B, Campbell JE. Targeting the incretin/glucagon system with triagonists to treat diabetes. Endocr Rev. 2018;39:519.

    Article  Google Scholar 

  44. Bhat VK, Kerr BD, Flatt PR, Gault VA. A novel GIP-oxyntomodulin hybrid peptide acting through GIP, glucagon and GLP-1 receptors exhibits weight reducing and anti-diabetic properties. Biochem Pharmacol. 2013.

  45. Gault VA, Bhat VK, Irwin N, Flatt PR. A novel glucagon-like peptide-1 (GLP-1)/glucagon hybrid peptide with triple-acting agonist activity at glucose-dependent insulinotropic polypeptide, GLP-1, and glucagon receptors and therapeutic potential in high fat-fed Mice. J Biol Chem. 2013.

  46. Bhat VK, Kerr BD, Vasu S, Flatt PR, Gault VA. A DPP-IV-resistant triple-acting agonist of GIP, GLP-1 and glucagon receptors with potent glucose-lowering and insulinotropic actions in high-fat-fed mice. Diabetologia. 2013.

  47. Deacon CF. Physiology and pharmacology of DPP-4 in glucose homeostasis and the treatment of type 2 diabetes. Front Endocrinol (Lausanne). 2019;10.

  48. Deacon CF. Dipeptidyl peptidase-4 inhibitors in the treatment of type 2 diabetes: a comparative review. Diabetes Obes Metab. 2011;13:7–18.

    Article  CAS  PubMed  Google Scholar 

  49. Craddy P, Palin H-J, Johnson KI. Comparative effectiveness of dipeptidylpeptidase-4 inhibitors in type 2 diabetes: a systematic review and mixed treatment comparison. Diabetes Ther. 2014;5:1–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Vella A, Bock G, Giesler PD, Burton DB, Serra DB, Saylan ML, et al. Effects of dipeptidyl peptidase-4 inhibition on gastrointestinal function, meal appearance, and glucose metabolism in type 2 diabetes. Diabetes. 2007;56:1475–80.

    Article  CAS  PubMed  Google Scholar 

  51. Athauda D, Foltynie T. Insulin resistance and Parkinson’s disease: a new target for disease modification? Prog Neurobiol. 2016;145:98–120.

    Article  PubMed  Google Scholar 

  52. Markaki I, Winther K, Catrina SB, Svenningsson P. Repurposing GLP1 agonists for neurodegenerative diseases. Int Rev Neurobiol. 2020.

  53. Glotfelty EJ, Olson L, Karlsson TE, Li Y, Greig NH. Glucagon-like peptide-1 (GLP-1)-based receptor agonists as a treatment for Parkinson’s disease. Expert Opin Investig Drugs. 2020;29:595–602.

    Article  CAS  PubMed  Google Scholar 

  54. DeTure MA, Dickson DW. The neuropathological diagnosis of Alzheimer’s disease. Mol Neurodegener. 2019;14:32.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Talbot K, Wang H, Kazi H, Han L, Bakshi KP, Stucky A, et al. Demonstrated brain insulin resistance in Alzheimer’s disease patients. J Clin Investig. 2012;122:1316–38.

    Article  CAS  PubMed  Google Scholar 

  56. Craft S, Raman R, Chow TW, Rafii MS, Sun C-K, Rissman RA, et al. Safety, efficacy, and feasibility of intranasal insulin for the treatment of mild cognitive impairment and Alzheimer disease dementia. JAMA Neurol. 2020;77:1099.

    Article  PubMed  Google Scholar 

  57. Craft S, Baker LD, Montine TJ, Minoshima S, Watson GS, Claxton A, et al. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Arch Neurol. 2012.

  58. Craft S, Claxton A, Baker LD, Hanson AJ, Cholerton B, Trittschuh EH, et al. Effects of regular and long-acting insulin on cognition and Alzheimer’s disease biomarkers: a pilot clinical trial. J Alzheimer’s Dis. 2017.

  59. Masciopinto F, Di Pietro N, Corona C, Bomba M, Pipino C, Curcio M, et al. Effects of long-term treatment with pioglitazone on cognition and glucose metabolism of PS1-KI, 3xTg-AD, and wild-type mice. Cell Death Dis. 2012;3:e448–e448.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Perry T, Lahiri DK, Sambamurti K, Chen D, Mattson MP, Egan JM, et al. Glucagon-like peptide-1 decreases endogenous amyloid-β peptide (Aβ) levels and protects hippocampal neurons from death induced by Aβ and iron. J Neurosci Res. 2003;72:603–12.

    Article  CAS  PubMed  Google Scholar 

  61. Cai H-Y, Yang J-T, Wang Z-J, Zhang J, Yang W, Wu M-N, et al. Lixisenatide reduces amyloid plaques, neurofibrillary tangles and neuroinflammation in an APP/PS1/tau mouse model of Alzheimer’s disease. Biochem Biophys Res Commun. 2018;495:1034–40.

    Article  CAS  PubMed  Google Scholar 

  62. Solmaz V, Çınar BP, Yiğittürk G, Çavuşoğlu T, Taşkıran D, Erbaş O. Exenatide reduces TNF-α expression and improves hippocampal neuron numbers and memory in streptozotocin treated rats. Eur J Pharmacol. 2015;765:482–7.

    Article  CAS  PubMed  Google Scholar 

  63. McClean PL, Hölscher C. Liraglutide can reverse memory impairment, synaptic loss and reduce plaque load in aged APP/PS1 mice, a model of Alzheimer’s disease. Neuropharmacology. 2014;76:57–67.

    Article  CAS  PubMed  Google Scholar 

  64. McClean PL, Jalewa J, Hölscher C. Prophylactic liraglutide treatment prevents amyloid plaque deposition, chronic inflammation and memory impairment in APP/PS1 mice. Behav Brain Res. 2015;293:96–106.

    Article  CAS  PubMed  Google Scholar 

  65. Hansen HH, Fabricius K, Barkholt P, Kongsbak-Wismann P, Schlumberger C, Jelsing J, et al. Long-term treatment with liraglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist, has no effect on β-amyloid plaque load in two transgenic APP/PS1 mouse models of Alzheimer’s disease. PLoS ONE. 2016;11:e0158205.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Cao Y, Hölscher C, Hu M-M, Wang T, Zhao F, Bai Y, et al. DA5-CH, a novel GLP-1/GIP dual agonist, effectively ameliorates the cognitive impairments and pathology in the APP/PS1 mouse model of Alzheimer’s disease. Eur J Pharmacol. 2018;827:215–26.

    Article  CAS  PubMed  Google Scholar 

  67. Shi L, Zhang Z, Li L, Hölscher C. A novel dual GLP-1/GIP receptor agonist alleviates cognitive decline by re-sensitizing insulin signaling in the Alzheimer ICV. STZ rat model. Behav Brain Res. 2017;327:65–74.

    Article  CAS  PubMed  Google Scholar 

  68. Tai J, Liu W, Li Y, Li L, Hölscher C. Neuroprotective effects of a triple GLP-1/GIP/glucagon receptor agonist in the APP/PS1 transgenic mouse model of Alzheimer’s disease. Brain Res. 2018;1678:64–74.

    Article  CAS  PubMed  Google Scholar 

  69. Salles GN, Calió ML, Hölscher C, Pacheco-Soares C, Porcionatto M, Lobo AO. Neuroprotective and restorative properties of the GLP-1/GIP dual agonist DA-JC1 compared with a GLP-1 single agonist in Alzheimer’s disease. Neuropharmacology. 2020;162:107813.

    Article  CAS  PubMed  Google Scholar 

  70. Mullins RJ, Mustapic M, Chia CW, Carlson O, Gulyani S, Tran J, et al. A pilot study of exenatide actions in Alzheimer’s disease. Curr Alzheimer Res. 2019;16:741–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Cairns E. CTAD 2020—Elad fails, but GLP-1s could still have a future in Alzheimer’s [Internet]. Evaluate. 2020. https://www.evaluate.com/vantage/articles/news/snippets/ctad-2020-elad-fails-glp-1s-could-still-have-future-alzheimers.

  72. Cukierman-Yaffe T, Gerstein HC, Colhoun HM, Diaz R, García-Pérez L-E, Lakshmanan M, et al. Effect of dulaglutide on cognitive impairment in type 2 diabetes: an exploratory analysis of the REWIND trial. Lancet Neurol. 2020;19:582–90.

    Article  CAS  PubMed  Google Scholar 

  73. Ballard C, Nørgaard CH, Friedrich S, Mørch LS, Gerds T, Møller DV, et al. Liraglutide and semaglutide: pooled post hoc analysis to evaluate risk of dementia in patients with type 2 diabetes. Alzheimer’s Dement. 2020;16:1–2.

    Article  Google Scholar 

  74. Kosaraju J, Gali CC, Khatwal RB, Dubala A, Chinni S, Holsinger RMD, et al. Saxagliptin: a dipeptidyl peptidase-4 inhibitor ameliorates streptozotocin induced Alzheimer’s disease. Neuropharmacology. 2013;72:291–300.

    Article  CAS  PubMed  Google Scholar 

  75. Chaudhuri KR, Naidu Y. Early Parkinson’s disease and non-motor issues. J Neurol. 2008;255:33–8.

    Article  PubMed  Google Scholar 

  76. Gibb WRG, Lees AJ. The relevance of the Lewy body to the pathogenesis of idiopathic Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1988;51:745–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Vijiaratnam N, Foltynie T. Therapeutic strategies to treat or prevent off episodes in adults with Parkinson’s disease. Drugs. 2020;80:775–96.

    Article  CAS  PubMed  Google Scholar 

  78. Athauda D, Foltynie T. The ongoing pursuit of neuroprotective therapies in Parkinson disease. Nat Rev Neurol. 2015;11:25–40.

    Article  CAS  PubMed  Google Scholar 

  79. Athauda D, Foltynie T. Drug repurposing in Parkinson’s disease. CNS Drugs. 2018;32:747–61.

    Article  CAS  PubMed  Google Scholar 

  80. Svenningsson P, Wirdefeldt K, Yin L, Fang F, Markaki I, Efendic S, et al. Reduced incidence of Parkinson’s disease after dipeptidyl peptidase-4 inhibitors—A nationwide case-control study. Mov Disord. 2016;31:1422–3.

    Article  PubMed  Google Scholar 

  81. Brauer R, Wei L, Ma T, Athauda D, Girges C, Vijiaratnam N, et al. Diabetes medications and risk of Parkinson’s disease: a cohort study of patients with diabetes. Brain. 2020;143:3067–76.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Harkavyi A, Abuirmeileh A, Lever R, Kingsbury AE, Biggs CS, Whitton PS. Glucagon-like peptide 1 receptor stimulation by exendin-4 reverses key deficits in distinct rodent models of Parkinson’s disease. J Neuroinflamm. 2008;5:19.

    Article  Google Scholar 

  83. Bertilsson G, Patrone C, Zachrisson O, Andersson A, Dannaeus K, Heidrich J, et al. Peptide hormone exendin-4 stimulates subventricular zone neurogenesis in the adult rodent brain and induces recovery in an animal model of Parkinson’s disease. J Neurosci Res. 2008;86:326–38.

    Article  CAS  PubMed  Google Scholar 

  84. Liu W, Jalewa J, Sharma M, Li G, Li L, Hölscher C. Neuroprotective effects of lixisenatide and liraglutide in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Neuroscience. 2015;303:42–50.

    Article  CAS  PubMed  Google Scholar 

  85. Kastin AJ, Akerstrom V. Entry of exendin-4 into brain is rapid but may be limited at high doses. Int J Obes. 2003;27:313–8.

    Article  CAS  Google Scholar 

  86. Badawi GA, Abd El Fattah MA, Zaki HF, El Sayed MI. Sitagliptin and liraglutide reversed nigrostriatal degeneration of rodent brain in rotenone-induced Parkinson’s disease. Inflammopharmacology. 2017;25:369–822.

    Article  CAS  PubMed  Google Scholar 

  87. Kim S, Moon M, Park S. Exendin-4 protects dopaminergic neurons by inhibition of microglial activation and matrix metalloproteinase-3 expression in an animal model of Parkinson’s disease. J Endocrinol. 2009;202:431–9.

    Article  CAS  PubMed  Google Scholar 

  88. Li Y, Perry T, Kindy MS, Harvey BK, Tweedie D, Holloway HW, et al. GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc Natl Acad Sci. 2009;106:1285–90.

    Article  CAS  PubMed  Google Scholar 

  89. Zhang L, Zhang L, Li L, Hölscher C. Neuroprotective effects of the novel GLP-1 long acting analogue semaglutide in the MPTP Parkinson’s disease mouse model. Neuropeptides. 2018;71:70–80.

    Article  CAS  PubMed  Google Scholar 

  90. Zhang L, Zhang L, Li L, Hölscher C. Semaglutide is neuroprotective and reduces α-synuclein levels in the chronic MPTP mouse model of Parkinson’s disease. J Parkinsons Dis. 2019;9:157–71.

    Article  PubMed  Google Scholar 

  91. Chung HK, Ho H-A, Pérez-Acuña D, Lee S-J. Modeling α-synuclein propagation with preformed fibril injections. J Mov Disord. 2019;12:139–51.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Yun SP, Kam T-I, Panicker N, Kim S, Oh Y, Park J-S, et al. Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson’s disease. Nat Med. 2018;24:931–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Yuan Z, Li D, Feng P, Xue G, Ji C, Li G, et al. A novel GLP-1/GIP dual agonist is more effective than liraglutide in reducing inflammation and enhancing GDNF release in the MPTP mouse model of Parkinson’s disease. Eur J Pharmacol. 2017;812:82–90.

    Article  CAS  PubMed  Google Scholar 

  94. Feng P, Zhang X, Li D, Ji C, Yuan Z, Wang R, et al. Two novel dual GLP-1/GIP receptor agonists are neuroprotective in the MPTP mouse model of Parkinson’s disease. Neuropharmacology. 2018;133:385–94.

    Article  CAS  PubMed  Google Scholar 

  95. Zhang L, Zhang L, Li Y, Li L, Melchiorsen JU, Rosenkilde M, et al. The novel dual GLP-1/GIP receptor agonist DA-CH5 is superior to single GLP-1 receptor agonists in the MPTP model of Parkinson’s disease. J Parkinsons Dis. 2020;10:523–42.

    Article  CAS  PubMed  Google Scholar 

  96. Liu W, Li Y, Jalewa J, Saunders-Wood T, Li L, Hölscher C. Neuroprotective effects of an oxyntomodulin analogue in the MPTP mouse model of Parkinson’s disease. Eur J Pharmacol. 2015;765:284–90.

    Article  CAS  PubMed  Google Scholar 

  97. Aviles-Olmos I, Dickson J, Kefalopoulou Z, Djamshidian A, Ell P, Soderlund T, et al. Exenatide and the treatment of patients with Parkinson’s disease. J Clin Investig. 2013;123:2730–6.

    Article  CAS  PubMed  Google Scholar 

  98. Aviles-Olmos I, Dickson J, Kefalopoulou Z, Djamshidian A, Kahan J, Ell P, et al. Motor and cognitive advantages persist 12 months after exenatide exposure in Parkinson’s disease. J Parkinsons Dis. 2015;4:337–44.

    Article  Google Scholar 

  99. Athauda D, Maclagan K, Skene SS, Bajwa-Joseph M, Letchford D, Chowdhury K, et al. Exenatide once weekly versus placebo in Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet. 2017;390:1664–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Athauda D, Gulyani S, Karnati HK, Li Y, Tweedie D, Mustapic M, et al. Utility of neuronal-derived exosomes to examine molecular mechanisms that affect motor function in patients with Parkinson disease: a secondary analysis of the exenatide-PD trial. JAMA Neurol. 2019;76:420–9.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Abdelsalam RM, Safar MM. Neuroprotective effects of vildagliptin in rat rotenone Parkinson’s disease model: role of RAGE-NFκB and Nrf2-antioxidant signaling pathways. J Neurochem. 2015;133:700–7.

    Article  CAS  PubMed  Google Scholar 

  102. Nassar NN, Al-Shorbagy MY, Arab HH, Abdallah DM. Saxagliptin: a novel anti-Parkinsonian approach. Neuropharmacology. 2015;89:308–17.

    Article  CAS  PubMed  Google Scholar 

  103. de Turnes JM, Bassani TB, Souza LC, Vital MABF. Ineffectiveness of saxagliptin as a neuroprotective drug in 6-OHDA-lesioned rats. J Pharm Pharmacol. 2018;70:1059–68.

    Article  PubMed  Google Scholar 

  104. Gilman S, Wenning GK, Low PA, Brooks DJ, Mathias CJ, Trojanowski JQ, et al. Second consensus statement on the diagnosis of multiple system atrophy. Neurology. 2008;71:670–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Papp MI, Kahn JE, Lantos PL. Glial cytoplasmic inclusions in the CNS of patients with multiple system atrophy (striatonigral degeneration, olivopontocerebellar atrophy and Shy-Drager syndrome). J Neurol Sci. 1989;94:79–100.

    Article  CAS  PubMed  Google Scholar 

  106. Pellecchia MT, Pivonello R, Longo K, Manfredi M, Tessitore A, Amboni M, et al. Multiple system atrophy is associated with changes in peripheral insulin-like growth factor system. Mov Disord. 2010;25:2621–6.

    Article  PubMed  Google Scholar 

  107. Ubhi K, Rockenstein E, Mante M, Inglis C, Adame A, Patrick C, et al. Neurodegeneration in a transgenic mouse model of multiple system atrophy is associated with altered expression of oligodendroglial-derived neurotrophic factors. J Neurosci. 2010;30:6236–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Bassil F, Canron MH, Vital A, Bezard E, Li Y, Greig NH, et al. Insulin resistance and exendin-4 treatment for multiple system atrophy. Brain. 2017;140:1420–36.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Sorli C, Harashima S, Tsoukas GM, Unger J, Karsbøl JD, Hansen T, et al. Efficacy and safety of once-weekly semaglutide monotherapy versus placebo in patients with type 2 diabetes (SUSTAIN 1): a double-blind, randomised, placebo-controlled, parallel-group, multinational, multicentre phase 3a trial. Lancet Diabetes Endocrinol. 2017;5:251–60.

    Article  CAS  PubMed  Google Scholar 

  110. Zinman B, Aroda VR, Buse JB, Cariou B, Harris SB, Hoff ST, et al. Efficacy, safety, and tolerability of oral semaglutide versus placebo added to insulin with or without metformin in patients with type 2 diabetes: the PIONEER 8 trial. Diabetes Care. 2019;42:2262–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Yamada Y, Katagiri H, Hamamoto Y, Deenadayalan S, Navarria A, Nishijima K, et al. Dose-response, efficacy, and safety of oral semaglutide monotherapy in Japanese patients with type 2 diabetes (PIONEER 9): a 52-week, phase 2/3a, randomised, controlled trial. Lancet Diabetes Endocrinol. 2020;8:377–91.

    Article  CAS  PubMed  Google Scholar 

  112. Sun F, Yu K, Yang Z, Wu S, Zhang Y, Shi L, et al. Impact of GLP-1 receptor agonists on major gastrointestinal disorders for type 2 diabetes mellitus: a mixed treatment comparison meta-analysis. Exp Diabetes Res. 2012;2012:1–14.

    Article  Google Scholar 

  113. Nauck MA, Kemmeries G, Holst JJ, Meier JJ. Rapid tachyphylaxis of the glucagon-like peptide 1-induced deceleration of gastric emptying in humans. Diabetes. 2011;60:1561–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Sharma JC, Vassallo M. Prognostic significance of weight changes in Parkinson’s disease: the Park–weight phenotype. Neurodegener Dis Manag. 2014;4:309–16.

    Article  PubMed  Google Scholar 

  115. Kostev K, Ouwens M, Grandy S, Johnsson KM, Qiao Q. Adherence to GLP-1 receptor agonist therapy administered by once-daily or once-weekly injection in patients with type 2 diabetes in Germany. Diabetes Metab Syndr Obes Targets Ther. 2016;9:201–5.

    Article  Google Scholar 

  116. Elashoff M, Matveyenko AV, Gier B, Elashoff R, Butler PC. Pancreatitis, pancreatic, and thyroid cancer with glucagon-like peptide-1-based therapies. Gastroenterology. 2011;141:150–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Vrang N, Jelsing J, Simonsen L, Jensen AE, Thorup I, Søeborg H, et al. The effects of 13 weeks of liraglutide treatment on endocrine and exocrine pancreas in male and female ZDF rats: a quantitative and qualitative analysis revealing no evidence of drug-induced pancreatitis. Am J Physiol Metab. 2012;303:E253–64.

    CAS  Google Scholar 

  118. Tatarkiewicz K, Belanger P, Gu G, Parkes D, Roy D. No evidence of drug-induced pancreatitis in rats treated with exenatide for 13 weeks. Diabetes Obes Metab. 2013;15:417–26.

    Article  CAS  PubMed  Google Scholar 

  119. Romley JA, Goldman DP, Solomon M, McFadden D, Peters AL. Exenatide therapy and the risk of pancreatitis and pancreatic cancer in a privately insured population. Diabetes Technol Ther. 2012;14:904–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Pinto LC, Falcetta MR, Rados DV, Leitão CB, Gross JL. Glucagon-like peptide-1 receptor agonists and pancreatic cancer: a meta-analysis with trial sequential analysis. Sci Rep. 2019;9:2375.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Gentilella R, Pechtner V, Corcos A, Consoli A. Glucagon-like peptide-1 receptor agonists in type 2 diabetes treatment: are they all the same? Diabetes Metab Res Rev. 2019;35:e3070.

    Article  PubMed  Google Scholar 

  122. Pinto LC, Rados DV, Barkan SS, Leitão CB, Gross JL. Dipeptidyl peptidase-4 inhibitors, pancreatic cancer and acute pancreatitis: a meta-analysis with trial sequential analysis. Sci Rep. 2018;8:782.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Nauck MA, Vilsboll T, Gallwitz B, Garber A, Madsbad S. Incretin-based therapies: viewpoints on the way to consensus. Diabetes Care. 2009;32:S223–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Onoviran OF, Li D, Toombs Smith S, Raji MA. Effects of glucagon-like peptide 1 receptor agonists on comorbidities in older patients with diabetes mellitus. Ther Adv Chronic Dis. 2019;10:204062231986269.

    Article  Google Scholar 

  125. Breen DP, Halliday GM, Lang AE. Gut–brain axis and the spread of α-synuclein pathology: vagal highway or dead end? Mov Disord. 2019;34:307–16.

    Article  PubMed  Google Scholar 

  126. Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, et al. Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell. 2016;167:1469–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Hunter K, Hölscher C. Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neurosci. 2012;13:33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Secher A, Jelsing J, Baquero AF, Hecksher-Sørensen J, Cowley MA, Dalbøge LS, et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J Clin Investig. 2014;124:4473–88.

    Article  CAS  PubMed  Google Scholar 

  129. Qin C, Li J, Tang K. The paraventricular nucleus of the hypothalamus: development, function, and human diseases. Endocrinology. 2018;159:3458–72.

    Article  CAS  PubMed  Google Scholar 

  130. Yin W, Gore AC. The hypothalamic median eminence and its role in reproductive aging. Ann N Y Acad Sci. 2010;1204:113–22.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Christensen M, Sparre-Ulrich AH, Hartmann B, Grevstad U, Rosenkilde MM, Holst JJ, et al. Transfer of liraglutide from blood to cerebrospinal fluid is minimal in patients with type 2 diabetes. Int J Obes. 2015;39:1651–4.

    Article  CAS  Google Scholar 

  132. Chen S, Yu SJ, Li Y, Lecca D, Glotfelty E, Kim HK, et al. Post-treatment with PT302, a long-acting exendin-4 sustained release formulation, reduces dopaminergic neurodegeneration in a 6-hydroxydopamine rat model of Parkinson’s disease. Sci Rep. 2018.

  133. Shan Y, Tan S, Lin Y, Liao S, Zhang B, Chen X, et al. The glucagon-like peptide-1 receptor agonist reduces inflammation and blood-brain barrier breakdown in an astrocyte-dependent manner in experimental stroke. J Neuroinflamm. 2019;16:242.

    Article  CAS  Google Scholar 

  134. Li C, Liu W, Li X, Zhang Z, Qi H, Liu S, et al. The novel GLP-1/GIP analogue DA5-CH reduces tau phosphorylation and normalizes theta rhythm in the icv. STZ rat model of AD. Brain Behav. 2020;10:e01505.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Gabery S, Salinas CG, Paulsen SJ, Ahnfelt-Rønne J, Alanentalo T, Baquero AF, et al. Semaglutide lowers body weight in rodents via distributed neural pathways. JCI Insight. 2020;5.

  136. Nauck MA, Stewart MW, Perkins C, Jones-Leone A, Yang F, Perry C, et al. Efficacy and safety of once-weekly GLP-1 receptor agonist albiglutide (HARMONY 2): 52 week primary endpoint results from a randomised, placebo-controlled trial in patients with type 2 diabetes mellitus inadequately controlled with diet and exercise. Diabetologia. 2016;59:266–74.

    Article  CAS  PubMed  Google Scholar 

  137. Salameh TS, Rhea EM, Talbot K, Banks WA. Brain uptake pharmacokinetics of incretin receptor agonists showing promise as Alzheimer’s and Parkinson’s disease therapeutics. Biochem Pharmacol. 2020;180:114187.

    Article  CAS  PubMed  Google Scholar 

  138. Chowen JA, de Fonseca FR, Alvarez E, Navarro M, Garcı́a-egura LM, Blázquez E. Increased glucagon-like peptide-1 receptor expression in glia after mechanical lesion of the rat brain. Neuropeptides. 1999;33:212–5.

    Article  CAS  PubMed  Google Scholar 

  139. Ayoub BM, Mowaka S, Safar MM, Ashoush N, Arafa MG, Michel HE, et al. Repositioning of Omarigliptin as a once-weekly intranasal anti-Parkinsonian Agent. Sci Rep. 2018;8:8959.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

None to declare.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Thomas Foltynie.

Ethics declarations

Funding

No funding was received specifically for the publication of this review.

Conflict of interest

CG reports no conflicts of interest. NV has received unconditional educational grants from IPSEN and Biogen, travel grants from IPSEN, AbbVie and The International Parkinson’s Disease and Movement Disorders Society, speaker’s honorarium from AbbVie and STADA and served on advisory boards for Abbvie and Brittania outside of the submitted work. DA has received travel grants from Bial. GA reports no conflicts of interest. SG reports no conflicts of interest. TF has received grants from National Institute of Health Research, Michael J Fox Foundation, John Black Charitable Foundation, Cure Parkinson’s Trust, Innovate UK, Van Andel Research Institute and Defeat MSA. He has served on Advisory Boards for Voyager Therapeutics, Handl therapeutics, Living Cell Technologies, Bial, and Profie Pharma. He has received honoraria for talks sponsored by Bial, Profile Pharma, and Boston Scientific.

Ethics approval

Not applicable.

Consent to participate

Not applicable.

Consent for publication

Not applicable.

Availability of data and material

Not applicable.

Code availability

Not applicable.

Authors’ Contributions

CG: Review of literature, preparation of manuscript, critical revision for intellectual content; NV: Interpretation of data, critical revision of manuscript for intellectual content; DA: Interpretation of data, critical revision of manuscript for intellectual content; GA: Critical revision of manuscript for intellectual content; SG: Critical revision of manuscript for intellectual content; TF: Article concept and design, interpretation of data, critical revision of manuscript for intellectual content.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Girges, C., Vijiaratnam, N., Athauda, D. et al. The Future of Incretin-Based Approaches for Neurodegenerative Diseases in Older Adults: Which to Choose? A Review of their Potential Efficacy and Suitability. Drugs Aging 38, 355–373 (2021). https://doi.org/10.1007/s40266-021-00853-7

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40266-021-00853-7

Navigation