open access

Vol 58, No 2 (2024)
Invited Review Article
Submitted: 2023-06-30
Accepted: 2023-07-25
Published online: 2023-08-22
Get Citation

Updates on pharmacological treatment for Alzheimer’s disease

Philip W. Tipton1
·
Pubmed: 37606550
·
Neurol Neurochir Pol 2024;58(2):150-160.
Affiliations
  1. Department of Neurology, Mayo Clinic, Jacksonville, Florida, United States

open access

Vol 58, No 2 (2024)
Invited review articles
Submitted: 2023-06-30
Accepted: 2023-07-25
Published online: 2023-08-22

Abstract

Alzheimer’s disease (AD) is the most common cause of dementia, and its rising prevalence is constantly increasing the global health burden. There are currently no curative therapies for AD, and current treatment options provide only modest clinical benefit. Despite numerous clinical trials, there have been no major additions to the AD treatment armamentarium this century. The prevailing pathomechanistic hypothesis for AD begins with abnormal accumulation of amyloid β (Aβ) leading to plaque development, and disease-modifying candidate therapies have largely aimed to disrupt this process. Numerous clinical trials of monoclonal antibodies directed at various stages of Aβ plaque development have yielded mostly negative results; however, recent results suggest that a breakthrough may be on the horizon. The past two years have yielded positive results for three monoclonal antibodies (aducanumab, lecanemab, and donanemab) although questions remain regarding their clinical effectiveness. Additional clarity is needed to determine whether the clinical benefits are great enough to offset the treatment risks and the resource implications for healthcare systems. This review provides a foundational context and update on recent disease-modifying therapies for AD that have reached Phase III clinical trials. Up-to-date information on these therapies will help clinicians better inform their clinical decision-making and the counselling they can offer patients and their carers.

Abstract

Alzheimer’s disease (AD) is the most common cause of dementia, and its rising prevalence is constantly increasing the global health burden. There are currently no curative therapies for AD, and current treatment options provide only modest clinical benefit. Despite numerous clinical trials, there have been no major additions to the AD treatment armamentarium this century. The prevailing pathomechanistic hypothesis for AD begins with abnormal accumulation of amyloid β (Aβ) leading to plaque development, and disease-modifying candidate therapies have largely aimed to disrupt this process. Numerous clinical trials of monoclonal antibodies directed at various stages of Aβ plaque development have yielded mostly negative results; however, recent results suggest that a breakthrough may be on the horizon. The past two years have yielded positive results for three monoclonal antibodies (aducanumab, lecanemab, and donanemab) although questions remain regarding their clinical effectiveness. Additional clarity is needed to determine whether the clinical benefits are great enough to offset the treatment risks and the resource implications for healthcare systems. This review provides a foundational context and update on recent disease-modifying therapies for AD that have reached Phase III clinical trials. Up-to-date information on these therapies will help clinicians better inform their clinical decision-making and the counselling they can offer patients and their carers.

Get Citation

Keywords

dementia, Alzheimer’s disease, treatment, amyloid, clinical trials

About this article
Title

Updates on pharmacological treatment for Alzheimer’s disease

Journal

Neurologia i Neurochirurgia Polska

Issue

Vol 58, No 2 (2024)

Article type

Invited Review Article

Pages

150-160

Published online

2023-08-22

Page views

1168

Article views/downloads

1063

DOI

10.5603/pjnns.96286

Pubmed

37606550

Bibliographic record

Neurol Neurochir Pol 2024;58(2):150-160.

Keywords

dementia
Alzheimer’s disease
treatment
amyloid
clinical trials

Authors

Philip W. Tipton

References (80)
  1. Rajan KB, Weuve J, Barnes LL, et al. Population estimate of people with clinical Alzheimer's disease and mild cognitive impairment in the United States (2020-2060). Alzheimers Dement. 2021; 17(12): 1966–1975.
  2. Li X, Feng X, Sun X, et al. Global, regional, and national burden of Alzheimer's disease and other dementias, 1990-2019. Front Aging Neurosci. 2022; 14: 937486.
  3. 2023 Alzheimer's disease facts and figures. Alzheimers Dement. 2023; 19(4): 1598–1695.
  4. Mokdad AH, Ballestros K, Echko M, et al. US Burden of Disease Collaborators. The State of US Health, 1990-2016: Burden of Diseases, Injuries, and Risk Factors Among US States. JAMA. 2018; 319(14): 1444–1472.
  5. Zissimopoulos J, Crimmins E, St Clair P. The Value of Delaying Alzheimer's Disease Onset. Forum Health Econ Policy. 2014; 18(1): 25–39.
  6. Brier MR, Gordon B, Friedrichsen K, et al. Tau and Aβ imaging, CSF measures, and cognition in Alzheimer's disease. Sci Transl Med. 2016; 8(338): 338ra66.
  7. Jack CR, Holtzman DM. Biomarker modeling of Alzheimer's disease. Neuron. 2013; 80(6): 1347–1358.
  8. Cummings J, Zhou Y, Lee G, et al. Alzheimer's disease drug development pipeline: 2023. Alzheimers Dement (N Y). 2023; 9(2): e12385.
  9. Makin S. The amyloid hypothesis on trial. Nature. 2018; 559(7715): S4–S7.
  10. Tipton PW, Bülbül N, Crook J, et al. Effects of sex and APOE on Parkinson's Disease-related cognitive decline. Neurol Neurochir Pol. 2021; 55(6): 559–566.
  11. Thal DR, Rüb U, Orantes M, et al. Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology. 2002; 58(12): 1791–1800.
  12. Sevigny J, Chiao P, Bussière T, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer's disease. Nature. 2016; 537(7618): 50–56.
  13. Sokołowska N, Sokołowski R, Oleksy E, et al. Usefulness of the Polish versions of the Montreal Cognitive Assessment 7.2 and the Mini-Mental State Examination as screening instruments for the detection of mild neurocognitive disorder. Neurol Neurochir Pol. 2020; 54(5): 440–448.
  14. Salloway S, Chalkias S, Barkhof F, et al. Amyloid-Related Imaging Abnormalities in 2 Phase 3 Studies Evaluating Aducanumab in Patients With Early Alzheimer Disease. JAMA Neurol. 2022; 79(1): 13–21.
  15. Honig LS, Vellas B, Woodward M, et al. Trial of Solanezumab for Mild Dementia Due to Alzheimer's Disease. N Engl J Med. 2018; 378(4): 321–330.
  16. Ostrowitzki S, Lasser RA, Dorflinger E, et al. SCarlet RoAD Investigators, SCarlet RoAD Investigators. A phase III randomized trial of gantenerumab in prodromal Alzheimer's disease. Alzheimers Res Ther. 2017; 9(1): 95.
  17. Salloway S, Honigberg LA, Cho W, et al. Amyloid positron emission tomography and cerebrospinal fluid results from a crenezumab anti-amyloid-beta antibody double-blind, placebo-controlled, randomized phase II study in mild-to-moderate Alzheimer's disease (BLAZE). Alzheimers Res Ther. 2018; 10(1): 96.
  18. Budd Haeberlein S, Aisen PS, Barkhof F, et al. Two Randomized Phase 3 Studies of Aducanumab in Early Alzheimer's Disease. J Prev Alzheimers Dis. 2022; 9(2): 197–210.
  19. Mafi JN, Leng M, Arbanas JC, et al. Estimated Annual Spending on Aducanumab in the US Medicare Program. JAMA Health Forum. 2022; 3(1): e214495.
  20. Safety and Feasibility of Exablate Blood-Brain Barrier Disruption for Mild Cognitive Impairment or Mild Alzheimer's Disease Undergoing Aduhelm Therapy. https://ClinicalTrials.gov/show/NCT05469009.
  21. Lowe SL, Willis BA, Hawdon A, et al. Donanemab (LY3002813) dose-escalation study in Alzheimer's disease. Alzheimers Dement (N Y). 2021; 7(1): e12112.
  22. Mintun MA, Lo AC, Duggan Evans C, et al. Donanemab in Early Alzheimer's Disease. N Engl J Med. 2021; 384(18): 1691–1704.
  23. Shcherbinin S, Evans CD, Lu M, et al. Association of Amyloid Reduction After Donanemab Treatment With Tau Pathology and Clinical Outcomes: The TRAILBLAZER-ALZ Randomized Clinical Trial. JAMA Neurol. 2022; 79(10): 1015–1024.
  24. Gueorguieva I, Willis BA, Chua L, et al. Donanemab Population Pharmacokinetics, Amyloid Plaque Reduction, and Safety in Participants with Alzheimer's Disease. Clin Pharmacol Ther. 2023; 113(6): 1258–1267.
  25. Pontecorvo MJ, Lu M, Burnham SC, et al. Association of Donanemab Treatment With Exploratory Plasma Biomarkers in Early Symptomatic Alzheimer Disease: A Secondary Analysis of the TRAILBLAZER-ALZ Randomized Clinical Trial. JAMA Neurol. 2022; 79(12): 1250–1259.
  26. A Follow-On Study of Donanemab (LY3002813) With Video Assessments in Participants With Alzheimer's Disease (TRAILBLAZER-EXT). https://ClinicalTrials.gov/show/NCT04640077.
  27. Sims JR, Zimmer JA, Evans CD, et al. TRAILBLAZER-ALZ 2 Investigators. Donanemab in Early Symptomatic Alzheimer Disease: The TRAILBLAZER-ALZ 2 Randomized Clinical Trial. JAMA. 2023 [Epub ahead of print].
  28. A Donanemab (LY3002813) Prevention Study in Participants With Alzheimer's Disease (TRAILBLAZER-ALZ 3). https://ClinicalTrials.gov/show/NCT05026866.
  29. A Study of Donanemab (LY3002813) Compared With Aducanumab in Participants With Early Symptomatic Alzheimer's Disease (TRAILBLAZER-ALZ 4). https://ClinicalTrials.gov/show/NCT05108922.
  30. A Study of Donanemab (LY3002813) in Participants With Early Symptomatic Alzheimer's Disease (TRAILBLAZER-ALZ 5). https://ClinicalTrials.gov/show/NCT05508789.
  31. Bateman RJ, Cummings J, Schobel S, et al. Gantenerumab: an anti-amyloid monoclonal antibody with potential disease-modifying effects in early Alzheimer's disease. Alzheimers Res Ther. 2022; 14(1): 178.
  32. Dubois B, Feldman HH, Jacova C, et al. Research criteria for the diagnosis of Alzheimer's disease: revising the NINCDS-ADRDA criteria. Lancet Neurol. 2007; 6(8): 734–746.
  33. Ostrowitzki S, Deptula D, Thurfjell L, et al. Mechanism of amyloid removal in patients with Alzheimer disease treated with gantenerumab. Arch Neurol. 2012; 69(2): 198–207.
  34. Salloway SP, Sperling R, Fox NC, et al. Bapineuzumab 301 and 302 Clinical Trial Investigators. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer's disease. N Engl J Med. 2014; 370(4): 322–333.
  35. Sperling R, Salloway S, Brooks DJ, et al. Amyloid-related imaging abnormalities in patients with Alzheimer's disease treated with bapineuzumab: a retrospective analysis. Lancet Neurol. 2012; 11(3): 241–249.
  36. Delor I, Charoin JE, Gieschke R, et al. Modeling Alzheimer's Disease Progression Using Disease Onset Time and Disease Trajectory Concepts Applied to CDR-SOB Scores From ADNI. CPT Pharmacometrics Syst Pharmacol. 2013; 2(10): e78.
  37. A Study of Gantenerumab in Participants With Prodromal Alzheimer's Disease. https://ClinicalTrials.gov/show/NCT01224106.
  38. A Study of Gantenerumab in Participants With Mild Alzheimer Disease. https://ClinicalTrials.gov/show/NCT02051608.
  39. Klein G, Delmar P, Kerchner GA, et al. Thirty-Six-Month Amyloid Positron Emission Tomography Results Show Continued Reduction in Amyloid Burden with Subcutaneous Gantenerumab. J Prev Alzheimers Dis. 2021; 8(1): 3–6.
  40. Klein G, Delmar P, Voyle N, et al. Gantenerumab reduces amyloid-β plaques in patients with prodromal to moderate Alzheimer's disease: a PET substudy interim analysis. Alzheimers Res Ther. 2019; 11(1): 101.
  41. Roche provides update on Phase III GRADUATE programme evaluating gantenerumab in early Alzheimer’s disease. 2022.
  42. A Study to Evaluate the Efficacy and Safety of Gantenerumab in Participants at Risk for or at the Earliest Stages of Alzheimer's Disease (AD). https://clinicaltrials.gov/show/NCT05256134.
  43. Dominantly Inherited Alzheimer Network Trial: An Opportunity to Prevent Dementia. A Study of Potential Disease Modifying Treatments in Individuals at Risk for or With a Type of Early Onset Alzheimer's Disease Caused by a Genetic Mutation. Master Protocol DIAN-TU-001. https://clinicaltrials.gov/show/NCT01760005.
  44. Salloway S, Farlow M, McDade E, et al. A trial of gantenerumab or solanezumab in dominantly inherited Alzheimer's disease. Nat Med. 2021; 27(7): 1187–1196.
  45. Doody RS, Thomas RG, Farlow M, et al. Alzheimer's Disease Cooperative Study Steering Committee, Solanezumab Study Group. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer's disease. N Engl J Med. 2014; 370(4): 311–321.
  46. Logovinsky V, Satlin A, Lai R, et al. Safety and tolerability of BAN2401--a clinical study in Alzheimer's disease with a protofibril selective Aβ antibody. Alzheimers Res Ther. 2016; 8(1): 14.
  47. Satlin A, Wang J, Logovinsky V, et al. Design of a Bayesian adaptive phase 2 proof-of-concept trial for BAN2401, a putative disease-modifying monoclonal antibody for the treatment of Alzheimer's disease. Alzheimers Dement (N Y). 2016; 2(1): 1–12.
  48. Wang J, Logovinsky V, Hendrix SB, et al. ADCOMS: a composite clinical outcome for prodromal Alzheimer's disease trials. J Neurol Neurosurg Psychiatry. 2016; 87(9): 993–999.
  49. Swanson CJ, Zhang Y, Dhadda S, et al. A randomized, double-blind, phase 2b proof-of-concept clinical trial in early Alzheimer's disease with lecanemab, an anti-Aβ protofibril antibody. Alzheimers Res Ther. 2021; 13(1): 80.
  50. McDade E, Cummings JL, Dhadda S, et al. Lecanemab in patients with early Alzheimer's disease: detailed results on biomarker, cognitive, and clinical effects from the randomized and open-label extension of the phase 2 proof-of-concept study. Alzheimers Res Ther. 2022; 14(1): 191.
  51. van Dyck CH, Swanson CJ, Aisen P, et al. Lecanemab in Early Alzheimer's Disease. N Engl J Med. 2023; 388(1): 9–21.
  52. Andrews JS, Desai U, Kirson NY, et al. Disease severity and minimal clinically important differences in clinical outcome assessments for Alzheimer's disease clinical trials. Alzheimers Dement (N Y). 2019; 5: 354–363.
  53. Lansdall CJ, McDougall F, Butler LM, et al. Establishing Clinically Meaningful Change on Outcome Assessments Frequently Used in Trials of Mild Cognitive Impairment Due to Alzheimer's Disease. J Prev Alzheimers Dis. 2023; 10(1): 9–18.
  54. Thambisetty M, Howard R. Lecanemab trial in AD brings hope but requires greater clarity. Nat Rev Neurol. 2023; 19(3): 132–133.
  55. Jönsson L, Wimo A, Handels R, et al. The affordability of lecanemab, an amyloid-targeting therapy for Alzheimer's disease: an EADC-EC viewpoint. Lancet Reg Health Eur. 2023; 29: 100657.
  56. Cummings J, Apostolova L, Rabinovici GD, et al. Lecanemab: Appropriate Use Recommendations. J Prev Alzheimers Dis. 2023; 10(3): 362–377.
  57. Wach-Klink A, Iżycka-Świeszewska E, Kozera G, et al. Cerebral microbleeds in neurological practice: concepts, diagnostics and clinical aspects. Neurol Neurochir Pol. 2021; 55(5): 450–461.
  58. Jönsson L, Wimo A, Handels R, et al. The affordability of lecanemab, an amyloid-targeting therapy for Alzheimer's disease: an EADC-EC viewpoint. Lancet Reg Health Eur. 2023; 29: 100657.
  59. Piller C. Report on trial death stokes Alzheimer's drug fears. Science. 2023; 380(6641): 122–123.
  60. AHEAD 3-45 Study: A Study to Evaluate Efficacy and Safety of Treatment With Lecanemab in Participants With Preclinical Alzheimer's Disease and Elevated Amyloid and Also in Participants With Early Preclinical Alzheimer's Disease and Intermediate Amyloid. https://clinicaltrials.gov/show/NCT04468659.
  61. A Study of LY3372993 in Participants With Alzheimer's Disease (AD) and Healthy Participants. https://clinicaltrials.gov/show/NCT04451408.
  62. Jin Y. Safety and amyloid plaque reduction effects of remternetug in patients with Alzheimer’s disease: interim analysis from a phase 1 study. AD/PD. Gothenburg, Sweden 2023.
  63. A Study of Remternetug (LY3372993) in Participants With Alzheimer's Disease (TRAILRUNNER-ALZ 1). https://clinicaltrials.gov/show/NCT05463731.
  64. Roberts M, Sevastou I, Imaizumi Y, et al. Pre-clinical characterisation of E2814, a high-affinity antibody targeting the microtubule-binding repeat domain of tau for passive immunotherapy in Alzheimer's disease. Acta Neuropathol Commun. 2020; 8(1): 13.
  65. A Study to Assess Safety, Tolerability, Pharmacokinetics (PK), Immunogenicity, and Pharmacodynamics (PD) of Intravenous Infusions of E2814 in Healthy Participants. https://clinicaltrials.gov/show/NCT04231513.
  66. Ji C, Sigurdsson EM. Current Status of Clinical Trials on Tau Immunotherapies. Drugs. 2021; 81(10): 1135–1152.
  67. Dominantly Inherited Alzheimer Network Trial: An Opportunity to Prevent Dementia. A Study of Potential Disease Modifying Treatments in Individuals With a Type of Early Onset Alzheimer's Disease Caused by a Genetic Mutation (DIAN-TU). https://clinicaltrials.gov/show/NCT05269394.
  68. Salloway SP, Sevingy J, Budur K, et al. Advancing combination therapy for Alzheimer's disease. Alzheimers Dement (N Y). 2020; 6(1): e12073.
  69. Middleton G, Silcocks P, Cox T, et al. Gemcitabine and capecitabine with or without telomerase peptide vaccine GV1001 in patients with locally advanced or metastatic pancreatic cancer (TeloVac): an open-label, randomised, phase 3 trial. Lancet Oncol. 2014; 15(8): 829–840.
  70. Park HH, Lee KY, Kim S, et al. Novel vaccine peptide GV1001 effectively blocks β-amyloid toxicity by mimicking the extra-telomeric functions of human telomerase reverse transcriptase. Neurobiol Aging. 2014; 35(6): 1255–1274.
  71. Park HH, Yu HJ, Kim S, et al. Neural stem cells injured by oxidative stress can be rejuvenated by GV1001, a novel peptide, through scavenging free radicals and enhancing survival signals. Neurotoxicology. 2016; 55: 131–141.
  72. Koh SH, Kwon HS, Choi SH, et al. Efficacy and safety of GV1001 in patients with moderate-to-severe Alzheimer's disease already receiving donepezil: a phase 2 randomized, double-blind, placebo-controlled, multicenter clinical trial. Alzheimers Res Ther. 2021; 13(1): 66.
  73. GV1001 Subcutaneous(SC) for the Treatment of Mild to Moderate Alzheimer's Disease (AD). https://clinicaltrials.gov/show/NCT05189210.
  74. GV1001 Subcutaneous for the Treatment of Moderate to Severe Alzheimer's Disease (AD). https://clinicaltrials.gov/show/NCT05303701.
  75. Bednarz K, Siuda J. Alzheimer's disease and type 2 diabetes mellitus: similarities in pathomechanisms lead to therapeutic opportunities. Neurol Neurochir Pol. 2021; 55(5): 418–428.
  76. During MJ, Cao L, Zuzga DS, et al. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat Med. 2003; 9(9): 1173–1179.
  77. Zhang ZQ, Hölscher C. Novel dual GLP-1/GIP receptor agonists show neuroprotective effects in Alzheimer's and Parkinson's disease models. Neuropharmacology. 2018; 136(Pt B): 251–259.
  78. Salameh TS, Rhea EM, Talbot K, et al. Brain uptake pharmacokinetics of incretin receptor agonists showing promise as Alzheimer's and Parkinson's disease therapeutics. Biochem Pharmacol. 2020; 180: 114187.
  79. A Research Study Investigating Semaglutide in People With Early Alzheimer's Disease (EVOKE). https://clinicaltrials.gov/show/NCT04777396.
  80. A Research Study Investigating Semaglutide in People With Early Alzheimer's Disease (EVOKE Plus). https://clinicaltrials.gov/show/NCT04777409.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp. z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk, Poland
tel.:+48 58 320 94 94, fax:+48 58 320 94 60, e-mail: viamedica@viamedica.pl