Volume 28, Issue 3 (September 2024)                   Physiol Pharmacol 2024, 28(3): 219-236 | Back to browse issues page


XML Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

Joodaki M, Merati Shirazi M, Hosseini N. New drugs for Alzheimer’s disease: Aducanumab or Donanemab?. Physiol Pharmacol 2024; 28 (3) : 1
URL: http://ppj.phypha.ir/article-1-2164-en.html
Abstract:   (1122 Views)

The main pathological features of Alzheimer’s disease (AD) include the cytotoxic extracellular accumulation of the amyloid beta (Aβ) plaques and intracellular neurofibrillary tangles. The Aβ plaques are responsible for cholinergic dysfunction and dementia in AD patients. Immunoglobulin G (IgG) and Aβ form an immune complex that activates neuroglia, clearing Aβ from the brain. Various Aβ-based therapeutic strategies have been proposed to reduce Aβ production, inhibit Aβ aggregation, and increase Aβ clearance. New medicines, such as aducanumab and donanemab, which are human IgG1 monoclonal antibodies, reduce cognitive impairment in patients with AD by decreasing the amount of Aβ plaques. Despite the considerable advantages of these agents, some disadvantages have also been reported, including Aβ-related imaging abnormalities, anaphylaxis, high cost, and contradictory results. Moreover, donanemab has delivered contradictory outcomes in improving recognition and performance in AD. However, although not fully proven yet, fewer side effects are reported for donanemab compared to aducanumab. Therefore, this review aims to explore the research background, compare the mechanism of action, and understand the advantages and disadvantages of aducanumab and donanemab. As a result, these medicines with maximum effectiveness and safety, yet fewer side effects, could be developed for future treatment and references.

Article number: 1
Full-Text [PDF 856 kb]   (152 Downloads)    
Type of Manuscript: Review | Subject: Neurophysiology/Pharmacology

References
1. Abyadeh M, Gupta V, Gupta V, Chitranshi N, Wu Y, Amirkhani A, et al. Comparative analysis of aducanumab, zagotenemab and pioglitazone as targeted treatment strategies for Alzheimer’s disease. Aging and disease 2021; 12: 1964. [DOI:10.14336/AD.2021.0719]
2. Aisen P S, Cummings J, Schneider L S. Symptomatic and nonamyloid/tau based pharmacologic treatment for Alzheimer disease. Cold spring harbor perspectives in medicine 2012; 2: a006395. [DOI:10.1101/cshperspect.a006395]
3. Arndt J W, Qian F, Smith B A, Quan C, Kilambi K P, Bush M W, et al. Structural and kinetic basis for the selectivity of aducanumab for aggregated forms of amyloid-β. Scientific reports 2018; 8: 1-16. [DOI:10.1038/s41598-018-24501-0]
4. Ayton S. Brain volume loss due to donanemab. Eur J Neurol 2021; 28: e67-e68. [DOI:10.1111/ene.15007]
5. Bartzokis G. Age-related myelin breakdown: a developmental model of cognitive decline and Alzheimer’s disease. Neurobiology of aging 2004; 25: 5-18. [DOI:10.1016/j.neurobiolaging.2003.03.001]
6. Beshir S A, Aadithsoorya A, Parveen A, Goh S S L, Hussain N, Menon V B. Aducanumab therapy to treat Alzheimer’s disease: a narrative review. International journal of alzheimer’s disease 2022; 2022. [DOI:10.1155/2022/9343514]
7. Bianchetti A, Trabucchi M. Clinical aspects of Alzheimer’s disease. Aging clinical and experimental research 2001; 13: 221-230. [DOI:10.1007/BF03351480]
8. Buchman A S, Wilson R S, Bienias J L, Shah R C, Evans D A, Bennett D A. Change in body mass index and risk of incident Alzheimer disease. Neurology 2005; 65: 892-897. [DOI:10.1212/01.wnl.0000176061.33817.90]
9. Budd Haeberlein S, Aisen P, Barkhof F, Chalkias S, Chen T, Cohen S, et al. Two randomized phase 3 studies of aducanumab in early Alzheimer’s disease. The Journal of prevention of alzheimer’s disease 2022a; 9: 197-210. [DOI:10.14283/jpad.2022.30]
10. Cable J, Holtzman D M, Hyman B T, Tansey M G, Colonna M, Kellis M, et al. Alternatives to amyloid for Alzheimer’s disease therapies-a symposium report. 2020. [DOI:10.1111/nyas.14371]
11. Chiao P, Bedell B J, Avants B, Zijdenbos A P, Grand’Maison M, O’Neill P, et al. Impact of reference and target region selection on amyloid PET SUV ratios in the phase 1b PRIME study of aducanumab. Journal of nuclear medicine 2019; 60: 100-106. [DOI:10.2967/jnumed.118.209130]
12. Chiong W, Tolchin B D, Bonnie R J, Busl K M, Cruz-Flores S, Epstein L G, et al. Decisions with patients and families regarding aducanumab in Alzheimer disease, with recommendations for consent: AAN position statement. Neurology 2022; 98: 154-159. [DOI:10.1212/WNL.0000000000013053]
13. Coerver K, Melissa M Y, D’Abreu A, Wasserman M, Nair K. Practical considerations in the administration of aducanumab for the neurologist. neurology: Clinical practice 2021. [DOI:10.1212/CPJ.0000000000001144]
14. Costa T, Cauda F. A bayesian reanalysis of the phase III aducanumab (ADU) trial. J Alzheimers Dis: 2022;87(3):1009-1012. [DOI:10.3233/JAD-220132]
15. Cummings J, Aisen P, Apostolova L, Atri A, Salloway S, Weiner M. Aducanumab: appropriate use recommendations. The journal of prevention of Alzheimer’s disease 2021a; 8: 398-410. [DOI:10.14283/jpad.2022.34]
16. Cummings J, Aisen P, Lemere C, Atri A, Sabbagh M, Salloway S. Aducanumab produced a clinically meaningful benefit in association with amyloid lowering. Alzheimer’s research & therapy 2021c; 13: 1-3. [DOI:10.1186/s13195-021-00838-z]
17. Cummings J, Osse A M L, Cammann D, Powell J, Chen J. Anti-amyloid monoclonal antibodies for the treatment of Alzheimer’s disease. BioDrugs 2024; 38: 5-22. [DOI:10.1007/s40259-023-00633-2]
18. Decourt B, Boumelhem F, Pope E D, Shi J, Mari Z, Sabbagh M N. Critical appraisal of amyloid lowering agents in AD. Current neurology and neuroscience reports 2021; 21: 1-10. [DOI:10.1007/s11910-021-01125-y]
19. Eric L. Ross; Marc S. Weinberg; Steven E. Arnold. Cost-effectiveness of aducanumab and donanemab for early Alzheimer disease in the US. JAMA neurology 2022. [DOI:10.1001/jamaneurol.2022.0315]
20. Farrer L A, Cupples L A, Haines J L, Hyman B, Kukull W A, Mayeux R, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease: a meta-analysis. Jama 1997; 278: 1349-1356. [DOI:10.1001/jama.1997.03550160069041]
21. Ferrero J, Williams L, Stella H, Leitermann K, Mikulskis A, O’Gorman J, et al. First-in-human, double-blind, placebo-controlled, single-dose escalation study of aducanumab (BIIB037) in mild-to-moderate Alzheimer’s disease. Alzheimer’s & Dementia: Translational research & clinical interventions 2016; 2: 169-176. [DOI:10.1016/j.trci.2016.06.002]
22. Fleisher A S, Lowe S L, Liu P, Shcherbinin S, Li L, Chua L, et al. SSignificant And sustained florbetapir F18 uptake reduction in patients with symptomatic Alzheimer’s disease with Ly3002813, A Β-amyloid plaque-specific antibody. . Alzheimer’s & dementia 2018; 14: P239-P240. [DOI:10.1016/j.jalz.2018.06.2378]
23. Frautschy S A, Yang F, Irrizarry M, Hyman B, Saido T, Hsiao K, et al. Microglial response to amyloid plaques in APPsw transgenic mice. The American journal of pathology 1998; 152: 307.
24. Frost C V, Zacharias M. From monomer to fibril: Abeta-amyloid binding to Aducanumab antibody studied by molecular dynamics simulation. Proteins 2020; 88: 1592-1606. [DOI:10.1002/prot.25978]
25. Gamage K K, Kumar S. Aducanumab therapy ameliorates calcium overload in a mouse model of Alzheimer’s disease. Journal of neuroscience 2017; 37: 4430-4432. [DOI:10.1523/JNEUROSCI.0420-17.2017]
26. Glymour M.M., Weuve J, Dufouil C, Mayeda E.R., Aduhelm, the newly approved medication for Alzheimer disease: what epidemiologists can learn and what epidemiology can offer. American Journal of Epidemiology 2022; 191(8): 1347-1351. [DOI:10.1093/aje/kwac063]
27. Greig S L. Memantine ER/donepezil: a review in Alzheimer’s disease. CNS drugs 2015; 29: 963-970. [DOI:10.1007/s40263-015-0287-2]
28. Gunawardena I P, Retinasamy T, Shaikh M F. Is Aducanumab for LMICs? Promises and challenges. Brain sciences 2021; 11: 1547. [DOI:10.3390/brainsci11111547]
29. Hassan N A, Alshamari A K, Hassan A A, Elharrif M G, Alhajri A M, Sattam M, et al. Advances on therapeutic strategies for Alzheimer’s disease: from medicinal plant to nanotechnology. Molecules 2022; 27: 4839. [DOI:10.3390/molecules27154839]
30. Hebert L, Bienias J, Aggarwal N, Wilson R, Bennett D, Shah R, et al. Change in risk of Alzheimer disease over time. Neurology 2010; 75: 786-791. [DOI:10.1212/WNL.0b013e3181f0754f]
31. Hensley K. Neuroinflammation in Alzheimer’s disease: mechanisms, pathologic consequences, and potential for therapeutic manipulation. Journal of Alzheimer’s disease 2010; 21: 1-14. [DOI:10.3233/JAD-2010-1414]
32. Hershey L A, Tarawneh R. Clinical efficacy, drug safety, and surrogate endpoints: has aducanumab met all of its expectations? Neurology 2021; 97: 517-518. [DOI:10.1212/WNL.0000000000012453]
33. Huang Y. Aβ-independent roles of apolipoprotein E4 in the pathogenesis of Alzheimer’s disease. Trends in molecular medicine 2010; 16: 287-294. [DOI:10.1016/j.molmed.2010.04.004]
34. Imbimbo B P, Ippati S, Watling M, Imbimbo C. Role of monomeric amyloid-β in cognitive performance in Alzheimer’s disease: Insights from clinical trials with secretase inhibitors and monoclonal antibodies. Pharmacological research 2023; 187: 106631. [DOI:10.1016/j.phrs.2022.106631]
35. Irizarry M C, Sims J R, Lowe S L, Nakano M, Hawdon A, Willis B A, et al. Safety, pharmacokinetics (Pk), and florbetapir F-18 positron emission tomography (Pet) after multiple dose administration of Ly3002813, AΒ-amyloid plaque-specific antibody, in Alzheimer’s disease (Ad). Alzheimer’s & dementia 2016a; 12: 352-353. [DOI:10.1016/j.jalz.2016.06.665]
36. Kastanenka K V, Bussiere T, Shakerdge N, Qian F, Weinreb P H, Rhodes K, et al. Immunotherapy with aducanumab restores calcium homeostasis in Tg2576 mice. Journal of neuroscience 2016; 36: 12549-12558. [DOI:10.1523/JNEUROSCI.2080-16.2016]
37. Kim D, Bae G H, Kim H Y, Jeon H, Kim K, Shin J, et al. Orally administered benzofuran derivative disaggregated Aβ plaques and oligomers in the brain of 5XFAD Alzheimer transgenic mouse. ACS chemical neuroscience 2020; 12: 99-108. [DOI:10.1021/acschemneuro.0c00606]
38. Kim H, Jeong W, Kwon J, Kim Y, Park E-C, Jang S-I. Association between depression and the risk of Alzheimer’s disease using the Korean national health insurance service-elderly cohort. Scientific reports 2021; 11: 22591. [DOI:10.1038/s41598-021-02201-6]
39. Knight E M, Kim S H, Kottwitz J C, Hatami A, Albay R, Suzuki A, et al. Effective anti-Alzheimer Aβ therapy involves depletion of specific Aβ oligomer subtypes. Neurology-neuroimmunology neuroinflammation 2016; 3. [DOI:10.1212/NXI.0000000000000237]
40. Knopman D S, Jones D T, Greicius M D. Failure to demonstrate efficacy of aducanumab: An analysis of the EMERGE and ENGAGE trials as reported by Biogen, December 2019. Alzheimer’s & dementia 2021; 17: 696-701. [DOI:10.1002/alz.12213]
41. Knopman D S, Perlmutter J S. Prescribing aducanumab in the face of meager efficacy and real risks. Neurology 2021; 97: 545-547. [DOI:10.1212/WNL.0000000000012452]
42. Leinenga G, Koh W K, Götz J. A comparative study of the effects of Aducanumab and scanning ultrasound on amyloid plaques and behavior in the APP23 mouse model of Alzheimer disease. Alzheimer’s research & therapy 2021; 13: 1-14. [DOI:10.1186/s13195-021-00809-4]
43. Li J, Wu X, Tan X, Wang S, Qu R, Wu X, et al. The efficacy and safety of anti-Aβ agents for delaying cognitive decline in Alzheimer’s disease: a meta-analysis. Frontiers in aging neuroscience 2023; 15. [DOI:10.3389/fnagi.2023.1257973]
44. Lin L, Hua F, Salinas C, Young C, Bussiere T, Apgar J F, et al. Quantitative systems pharmacology model for Alzheimer’s disease to predict effect of aducanumab on brain amyloid CPT: Pharmacometrics & systems pharmacology 2022. [DOI:10.1002/psp4.12759]
45. Lowe S, Duggan Evans C, Shcherbinin S, Cheng Y-J, Willis B, Gueorguieva I, et al. Donanemab (LY3002813) phase 1b study in Alzheimer’s disease: rapid and sustained reduction of brain amyloid measured by florbetapir F18 imaging. The Journal of prevention of Alzheimer’s disease 2021a; 8: 414-424. [DOI:10.14283/jpad.2021.56]
46. Lowe S L, Willis B A, Hawdon A, Natanegara F, Chua L, Foster J, et al. Donanemab (LY3002813) dose-escalation study in Alzheimer’s disease. Alzheimer’s & Dementia 2021b; 7: e12112. [DOI:10.1002/trc2.12112]
47. Mintun M A, Lo A C, Duggan Evans C, Wessels A M, Ardayfio P A, Andersen S W, et al. Donanemab in early Alzheimer’s disease. New England Journal of Medicine 2021a; 384: 1691-1704. [DOI:10.1056/NEJMoa2100708]
48. Mintun M A, Wessels A M, Sims J R. Donanemab in early Alzheimer’s disease. Reply. The new england journal of medicine 2021b; 385: 667-667. [DOI:10.1056/NEJMc2109455]
49. Mo J J, Li J y, Yang Z, Liu Z, Feng J S. Efficacy and safety of anti-amyloid-β immunotherapy for Alzheimer’s disease: a systematic review and network meta-analysis. Annals of clinical and translational neurology 2017; 4: 931-942. [DOI:10.1002/acn3.469]
50. Pais M, Martinez L, Ribeiro O, Loureiro J, Fernandez R, Valiengo L, et al. Early diagnosis and treatment of Alzheimer’s disease: new definitions and challenges. Brazilian journal of psychiatry 2020; 42: 431-441. [DOI:10.1590/1516-4446-2019-0735]
51. Panza F, Lozupone M, Dibello V, Greco A, Daniele A, Seripa D, et al. Are antibodies directed against amyloid-β (Aβ) oligomers the last call for the Aβ hypothesis of Alzheimer’s disease? Immunotherapy 2019; 11: 3-6. [DOI:10.2217/imt-2018-0119]
52. Panza F, Seripa D, Solfrizzi V, Imbimbo B P, Lozupone M, Leo A, et al. Emerging drugs to reduce abnormal β-amyloid protein in Alzheimer’s disease patients. Expert opinion on emerging drugs 2016; 21: 377-391. [DOI:10.1080/14728214.2016.1241232]
53. Paul S, Planque S, Nishiyama Y. Immunological origin and functional properties of catalytic autoantibodies to amyloid β peptide. Journal of clinical immunology 2010; 30: 43-49. [DOI:10.1007/s10875-010-9414-5]
54. Petch J, Bressington D. Aducanumab for Alzheimer’s disease: The never-ending story that nurses should know. Nursing open 2021; 8: 1524. [DOI:10.1002/nop2.878]
55. Pritam Das M P M, Linda H. Younkin, Steven.G. Younkin, TTodd Eliot Golde. Reduced effectiveness of Abeta1-42 immunization in APP transgenic mice with significant amyloid deposition. Neurobiology of aging 2001; 22: 721-727. [DOI:10.1016/S0197-4580(01)00245-7]
56. Qiu C, Kivipelto M, Von Strauss E. Epidemiology of Alzheimer’s disease: occurrence, determinants, and strategies toward intervention. Dialogues in clinical neuroscience 2009; 11: 111. [DOI:10.31887/DCNS.2009.11.2/cqiu]
57. Ramanan V K, Day G S. Anti-amyloid therapies for Alzheimer disease: Finally, good news for patients. Molecular neurodegeneration 2023; 18: 42. [DOI:10.1186/s13024-023-00637-0]
58. Rashad A, Rasool A, Shaheryar M, Sarfraz A, Sarfraz Z, Robles-Velasco K, et al. Donanemab for Alzheimer’s disease: a systematic review of clinical trials. Healthcare 2022; 11: 32. [DOI:10.3390/healthcare11010032]
59. Refolo L M, Pappolla M A, Malester B, LaFrancois J, Bryant-Thomas T, Wang R, et al. Hypercholesterolemia accelerates the Alzheimer’s amyloid pathology in a transgenic mouse model. Neurobiology of disease 2000; 7: 321-331. [DOI:10.1006/nbdi.2000.0366]
60. Retinasamy T, Shaikh M F. Aducanumab for Alzheimer’s disease: an update. Neuroscience research notes 2021; 4: 17-20. [DOI:10.31117/neuroscirn.v4i2.81]
61. Rostagno A, Cabrera E, Lashley T, Ghiso J. N-terminally truncated Aβ4-x proteoforms and their relevance for Alzheimer’s pathophysiology. Translational neurodegeneration 2022; 11: 1-18. [DOI:10.1186/s40035-022-00303-3]
62. Rubio-Perez J M, Morillas-Ruiz J M. A review: inflammatory process in Alzheimer’s disease, role of cytokines. The scientific world journal 2012; 2012. [DOI:10.1100/2012/756357]
63. Ryman J T, Meibohm B. Pharmacokinetics of monoclonal antibodies. CPT: pharmacometrics & systems pharmacology 2017; 6: 576-588. [DOI:10.1002/psp4.12224]
64. Sabbagh M N, Cummings J. Open Peer Commentary to “Failure to demonstrate efficacy of aducanumab: An analysis of the EMERGE and ENGAGE Trials as reported by Biogen December 2019”. Alzheimer’s & dementia 2021; 17: 702. [DOI:10.1002/alz.12235]
65. Sahni J K, Doggui S, Ali J, Baboota S, Dao L, Ramassamy C. Neurotherapeutic applications of nanoparticles in Alzheimer’s disease. Journal of controlled release 2011; 152: 208-231. [DOI:10.1016/j.jconrel.2010.11.033]
66. Sáiz-Vázquez O, Gracia-García P, Ubillos-Landa S, Puente-Martínez A, Casado-Yusta S, Olaya B, et al. Depression as a risk factor for Alzheimer’s disease: a systematic review of longitudinal meta-analyses. Journal of clinical medicine 2021; 10: 1809. [DOI:10.3390/jcm10091809]
67. Salloway S, Chalkias S, Barkhof F, Burkett P, Barakos J, Purcell D, et al. Amyloid-related imaging abnormalities in 2 phase 3 studies evaluating aducanumab in patients with early Alzheimer disease. JAMA neurology 2022; 79: 13-21. [DOI:10.1001/jamaneurol.2021.4161]
68. Scearce-Levie K, Sanchez P E, Lewcock J W. Leveraging preclinical models for the development of Alzheimer disease therapeutics. Nature reviews drug discovery 2020; 19: 447-462. [DOI:10.1038/s41573-020-0065-9]
69. Sevigny J, Chiao P, Bussière T, Weinreb P H, Williams L, Maier M, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 2016; 537: 50-56. [DOI:10.1038/nature19323]
70. Silva M V F, Loures C d M G, Alves L C V, de Souza L C, Borges K B G, Carvalho M d G. Alzheimer’s disease: risk factors and potentially protective measures. Journal of biomedical science 2019; 26: 1-11. [DOI:10.1186/s12929-019-0524-y]
71. Sun B-L, Li W-W, Zhu C, Jin W-S, Zeng F, Liu Y-H, et al. Clinical research on Alzheimer’s disease: progress and perspectives. Neuroscience bulletin 2018; 34: 1111-1118. [DOI:10.1007/s12264-018-0249-z]
72. Synnott P G, Whittington M D, Lin G A, Rind D M, Pearson S D. The effectiveness and value of aducanumab for Alzheimer’s disease: a summary from the institute for clinical and economic review’s California technology assessment forum. Journal of managed care & specialty pharmacy 2021; 27: 1613-1617. [DOI:10.18553/jmcp.2021.27.11.1613]
73. Tabassum S, Misrani A, Yang L. Exploiting common aspects of obesity and Alzheimer’s disease. Frontiers in human neuroscience 2020; 14: 602360. [DOI:10.3389/fnhum.2020.602360]
74. Tampi R R, Forester B P, Agronin M. Aducanumab: evidence from clinical trial data and controversies. Drugs in context 2021; 10. [DOI:10.7573/dic.2021-7-3]
75. Thomas E, Wasunna-Smith B, Kuruvilla T. Aducanumab and disease modifying treatments for Alzheimer’s disease. Progress in neurology and psychiatry 2021; 25: 4-6. [DOI:10.1002/pnp.711]
76. Tolar M, Abushakra S, Hey J A, Porsteinsson A, Sabbagh M. Aducanumab, gantenerumab, BAN2401, and ALZ-801-the first wave of amyloid-targeting drugs for Alzheimer’s disease with potential for near term approval. Alzheimer’s research & therapy 2020; 12: 1-10. [DOI:10.1186/s13195-020-00663-w]
77. Tolar M, Hey J, Power A, Abushakra S. Neurotoxic soluble amyloid oligomers drive Alzheimer’s pathogenesis and represent a clinically validated target for slowing disease progression. International journal of molecular sciences 2021; 22: 6355. [DOI:10.3390/ijms22126355]
78. Touchon J, Lachaine J, Beauchemin C, Granghaud A, Rive B, Bineau S. The impact of memantine in combination with acetylcholinesterase inhibitors on admission of patients with Alzheimer’s disease to nursing homes: cost-effectiveness analysis in France. The european journal of health economics 2014; 15: 791-800. [DOI:10.1007/s10198-013-0523-y]
79. van der Kant R, Goldstein L S, Ossenkoppele R. Amyloid-β-independent regulators of tau pathology in Alzheimer disease. Nature reviews neuroscience 2020; 21: 21-35. [DOI:10.1038/s41583-019-0240-3]
80. Van Eldik L J, Carrillo M C, Cole P E, Feuerbach D, Greenberg B D, Hendrix J A, et al. The roles of inflammation and immune mechanisms in Alzheimer’s disease. Alzheimer’s & dementia: 2016; 2: 99-109. [DOI:10.1016/j.trci.2016.05.001]
81. Vaz M, Silvestre S. Alzheimer’s disease: recent treatment strategies. European journal of pharmacology 2020; 887: 173554. [DOI:10.1016/j.ejphar.2020.173554]
82. Vellas B J. The Geriatrician, the primary care physician, aducanumab and the FDA decision: from frustration to new hope. The journal of nutrition, health & aging 2021; 25: 821-823. [DOI:10.1007/s12603-021-1657-8]
83. Weller J, Budson A. Current understanding of Alzheimer’s disease diagnosis and treatment. F1000Research 2018; 7. [DOI:10.12688/f1000research.14506.1]
84. Wessels A, Siemers E, Yu P, Andersen S, Holdridge K, Sims J, et al. A combined measure of cognition and function for clinical trials: The integrated Alzheimer’s Disease Rating Scale (iADRS). The journal of prevention of Alzheimer’s disease 2015; 2: 227. [DOI:10.14283/jpad.2015.82]
85. Xu X, Li Y, Wang J, Cao Y, Feng C, Guo Y, et al. Family history of AD/Dementia, polygenic risk score for AD, and Parkinson’s disease. Movement Disorders Clinical Practice 2023; 10: 1787-1794. [DOI:10.1002/mdc3.13919]
86. Yang P, Sun F. Aducanumab: The first targeted Alzheimer’s therapy. Drug discoveries & therapeutics 2021; 15: 166-168. [DOI:10.5582/ddt.2021.01061]
87. Yiannopoulou K G, Anastasiou A I, Zachariou V, Pelidou S-H. Reasons for failed trials of disease-modifying treatments for Alzheimer disease and their contribution in recent research. Biomedicines 2019; 7: 97. [DOI:10.3390/biomedicines7040097]
88. Yona Levites P D, Robert W Price, Marjorie J Rochette, Lisa A Kostura, Eileen M McGowan, Michael P Murphy, et al. Anti-Abeta42- and anti-Abeta40-specific mAbs attenuate amyloid deposition in an Alzheimer disease mouse model. Journal of clinical investigation 2006; 116: 193-201. [DOI:10.1172/JCI25410]
89. Zimmer J, Solomon P, Evans C D, Lu M, Sims J R, Brooks D A, et al. TRAILBLAZER-ALZ 2: A phase 3 study to assess safety and efficacy of donanemab in early symptomatic Alzheimer’s disease (P18-3.005). Journal 2022. [DOI:10.1212/WNL.98.18_supplement.1688]

Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.