Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter March 18, 2024

Alzheimer’s disease and clinical trials

  • Deepraj Paul ORCID logo EMAIL logo , Rohini Agrawal ORCID logo and Swati Singh ORCID logo

Abstract

Alzheimer’s disease (AD) is spreading its root disproportionately among the worldwide population. Many genes have been identified as the hallmarks of AD. Based upon the knowledge, many clinical trials have been designed and conducted. Attempts have been made to alleviate the pathology associated with AD by targeting the molecular products of these genes. Irrespective of the understanding on the genetic component of AD, many clinical trials have failed and imposed greater challenges on the path of drug discovery. Therefore, this review aims to identify research and review articles to pinpoint the limitations of drug candidates (thiethylperazine, CT1812, crenezumab, CNP520, and lecanemab), which are under or withdrawn from clinical trials. Thorough analysis of the cross-talk pathways led to the identification of many confounding factors, which could interfere with the success of clinical trials with drug candidates such as thiethylperazine, CT1812, crenezumab, and CNP520. Though these drug candidates were enrolled in clinical trials, yet literature review shows many limitations. These limitations raise many questions on the rationale behind the enrollments of these drug candidates in clinical trials. A meticulous prior assessment of the outcome of clinical studies may stop risky clinical trials at their inceptions. This may save time, money, and resources.


Corresponding author: Deepraj Paul, Department of Pharmacology, College of Pharmacy JSS Academy of Technical Education, Noida, Uttar Pradesh, India, Phone: 91 9986492608, E-mail:

Acknowledgments

Authors are thankful to MedChemExpress for the chemical structures of thiethylperazine, CT1812, and CNP520.

  1. Research ethics: Not applicable.

  2. Informed consent: Not applicable.

  3. Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  4. Competing interests: Authors state no conflict of interest.

  5. Research funding: None.

  6. Data availability: Not Applicable.

References

1. Piaceri, I, Nacmias, B, Sorbi, S. Genetics of familial and sporadic Alzheimer’s disease. Front Biosci (Elite Ed) 2013;5:167–77. https://doi.org/10.2741/e605.Search in Google Scholar PubMed

2. Julia, TCW, Goate, AM. Genetics of β-amyloid precursor protein in Alzheimer’s disease. Cold Spring Harb Perspect Med 2017;7:a024539. https://doi.org/10.1101/cshperspect.a024539.Search in Google Scholar PubMed PubMed Central

3. Tanzi, RE. The genetics of Alzheimer disease. Cold Spring Harb Perspect Med 2012;2:a006296. https://doi.org/10.1101/cshperspect.a006296.Search in Google Scholar PubMed PubMed Central

4. Sleegers, K, Brouwers, N, Gijselinck, I, Theuns, J, Goossens, D, Wauters, J, et al.. APP duplication is sufficient to cause early onset Alzheimer’s dementia with cerebral amyloid angiopathy. Brain 2006;129:2977–83. https://doi.org/10.1093/brain/awl203.Search in Google Scholar PubMed

5. Blom, ES, Viswanathan, J, Kilander, L, Helisalmi, S, Soininen, H, Lannfelt, L, et al.. Low prevalence of APP duplications in Swedish and Finnish patients with early-onset Alzheimer’s disease. Eur J Hum Genet 2008;16:171–5. https://doi.org/10.1038/sj.ejhg.5201966.Search in Google Scholar PubMed

6. Thonberg, H, Fallström, M, Björkström, J, Schoumans, J, Nennesmo, I, Graff, C. Mutation screening of patients with Alzheimer disease identifies APP locus duplication in a Swedish patient. BMC Res Notes 2011;4:1–9. https://doi.org/10.1186/1756-0500-4-476.Search in Google Scholar PubMed PubMed Central

7. Hooli, BV, Mohapatra, G, Mattheisen, M, Parrado, AR, Roehr, JT, Shen, Y, et al.. Role of common and rare APP DNA sequence variants in Alzheimer disease. Neurology 2012;78:1250–7. https://doi.org/10.1212/wnl.0b013e3182515972.Search in Google Scholar

8. Nicolas, M, Hassan, BA. Amyloid precursor protein and neural development. Development 2014;141:2543–8. https://doi.org/10.1242/dev.108712.Search in Google Scholar PubMed

9. Chen, GF, Xu, TH, Yan, Y, Zhou, YR, Jiang, Y, Melcher, K, et al.. Amyloid beta: structure, biology and structure-based therapeutic development. Acta Pharmacol Sin 2017;38:1205–35. https://doi.org/10.1038/aps.2017.28.Search in Google Scholar PubMed PubMed Central

10. Chow, VW, Mattson, MP, Wong, PC, Gleichmann, M. An overview of APP processing enzymes and products. NeuroMolecular Med 2010;12:1–2. https://doi.org/10.1007/s12017-009-8104-z.Search in Google Scholar PubMed PubMed Central

11. Sandebring, A, Welander, H, Winblad, B, Graff, C, Tjernberg, LO. The pathogenic Aβ43 is enriched in familial and sporadic Alzheimer disease. PLoS One 2013;8:e55847. https://doi.org/10.1371/journal.pone.0055847.Search in Google Scholar PubMed PubMed Central

12. Pope, CA, Wilkins, HM, Swerdlow, RH, Wolfe, MS. Mutations in the amyloid-β protein precursor reduce mitochondrial function and alter gene expression independent of 42-residue amyloid-β peptide. J Alzheimers Dis 2021;83:1039–49. https://doi.org/10.3233/jad-210366.Search in Google Scholar PubMed PubMed Central

13. Paasila, PJ, Aramideh, JA, Sutherland, GT, Graeber, MB. Synapses, microglia, and lipids in Alzheimer’s disease. Front Neurosci 2022;15:778822. https://doi.org/10.3389/fnins.2021.778822.Search in Google Scholar PubMed PubMed Central

14. Scheuner, D, Eckman, C, Jensen, M, Song, X, Citron, M, Suzuki, N, et al.. Secreted amyloid β–protein similar to that in the senile plaques of Alzheimer’s disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer’s disease. Nat Med 1996;2:864–70. https://doi.org/10.1038/nm0896-864.Search in Google Scholar PubMed

15. Duering, M, Grimm, MO, Grimm, HS, Schröder, J, Hartmann, T. Mean age of onset in familial Alzheimer’s disease is determined by amyloid beta 42. Neurobiol Aging 2005;26:785–8. https://doi.org/10.1016/j.neurobiolaging.2004.08.002.Search in Google Scholar PubMed

16. Misrani, A, Tabassum, S, Yang, L. Mitochondrial dysfunction and oxidative stress in Alzheimer’s disease. Front Aging Neurosci 2021;13:57. https://doi.org/10.3389/fnagi.2021.617588.Search in Google Scholar PubMed PubMed Central

17. Spies, PE, Verbeek, MM, van Groen, T, Claassen, JA. Reviewing reasons for the decreased CSF Abeta42 concentration in Alzheimer disease. Front Biosci 2012;17:2024–34. https://doi.org/10.2741/4035.Search in Google Scholar PubMed

18. Sarasija, S, Laboy, JT, Ashkavand, Z, Bonner, J, Tang, Y, Norman, KR. Presenilin mutations deregulate mitochondrial Ca2+ homeostasis and metabolic activity causing neurodegeneration in Caenorhabditis elegans. Elife 2018;7:e33052. https://doi.org/10.7554/elife.33052.Search in Google Scholar

19. Bai, XC, Yan, C, Yang, G, Lu, P, Ma, D, Sun, L, et al.. An atomic structure of human γ-secretase. Nature 2015;525:212–7. https://doi.org/10.1038/nature14892.Search in Google Scholar PubMed PubMed Central

20. Bentahir, M, Nyabi, O, Verhamme, J, Tolia, A, Horré, K, Wiltfang, J, et al.. Presenilin clinical mutations can affect γ-secretase activity by different mechanisms. J Neurochem 2006;96:732–42. https://doi.org/10.1111/j.1471-4159.2005.03578.x.Search in Google Scholar PubMed

21. Aguilar, LR, Acosta-Uribe, J, Giraldo, MM, Moreno, S, Baena, A, Alzate, D, et al.. Genetic origin of a large family with a novel PSEN1 mutation (Ile416Thr). Alzheimers Dement 2019;15:709–19. https://doi.org/10.1016/j.jalz.2018.12.010.Search in Google Scholar PubMed PubMed Central

22. Zhang, S, Cai, F, Wu, Y, Bozorgmehr, T, Wang, Z, Zhang, SI, et al.. A presenilin-1 mutation causes Alzheimer disease without affecting Notch signaling. Mol Psychiatry 2020;25:603–13. https://doi.org/10.1038/s41380-018-0101-x.Search in Google Scholar PubMed

23. Acosta-Baena, N, Sepulveda-Falla, D, Lopera-Gómez, CM, Jaramillo-Elorza, MC, Moreno, S, Aguirre-Acevedo, DC, et al.. Pre-dementia clinical stages in presenilin 1 E280A familial early-onset Alzheimer’s disease: a retrospective cohort study. Lancet Neurol 2011;10:213–20. https://doi.org/10.1016/s1474-4422(10)70323-9.Search in Google Scholar PubMed

24. De Strooper, B, Karran, E. The cellular phase of Alzheimer’s disease. Cell 2016;164:603–15. https://doi.org/10.1016/j.cell.2015.12.056.Search in Google Scholar PubMed

25. Arboleda-Velasquez, JF, Lopera, F, O’Hare, M, Delgado-Tirado, S, Marino, C, Chmielewska, N, et al.. Resistance to autosomal dominant Alzheimer’s disease in an APOE3 Christchurch homozygote: a case report. Nat Med 2019;25:1680–3. https://doi.org/10.1038/s41591-019-0611-3.Search in Google Scholar PubMed PubMed Central

26. Mutoh, T, Kawamura, N, Hirabayashi, Y, Shima, S, Miyashita, T, Ito, S, et al.. Abnormal cross-talk between mutant presenilin 1 (I143T, G384A) and glycosphingolipid biosynthesis. FASEB J 2012;26:3065–74. https://doi.org/10.1096/fj.11-198630.Search in Google Scholar PubMed

27. Dodge, JC, Tamsett, TJ, Treleaven, CM, Taksir, TV, Piepenhagen, P, Sardi, SP, et al.. Glucosylceramide synthase inhibition reduces ganglioside GM3 accumulation, alleviates amyloid neuropathology, and stabilizes remote contextual memory in a mouse model of Alzheimer’s disease. Alzheimer’s Res Ther 2022;14:1–2. https://doi.org/10.1186/s13195-022-00966-0.Search in Google Scholar PubMed PubMed Central

28. Nagy, ZS, Esiri, MM, Jobst, KA, Johnston, C, Litchfield, S, Sim, E, et al.. Influence of the apolipoprotein E genotype on amyloid deposition and neurofibrillary tangle formation in Alzheimer’s disease. Neuroscience 1995;69:757–61. https://doi.org/10.1016/0306-4522(95)00331-c.Search in Google Scholar PubMed

29. Troutwine, BR, Hamid, L, Lysaker, CR, Strope, TA, Wilkins, HM. Apolipoprotein E and Alzheimer’s disease. Acta Pharm Sin B 2022;12:496–510. https://doi.org/10.1016/j.apsb.2021.10.002.Search in Google Scholar PubMed PubMed Central

30. Bekris, LM, Yu, CE, Bird, TD, Tsuang, DW. Genetics of Alzheimer disease. J Geriatr Psychiatry Neurol 2010;23:213–27. https://doi.org/10.1177/0891988710383571.Search in Google Scholar PubMed PubMed Central

31. Gharbi-Meliani, A, Dugravot, A, Sabia, S, Regy, M, Fayosse, A, Schnitzler, A, et al.. The association of APOE ε4 with cognitive function over the adult life course and incidence of dementia: 20 years follow-up of the Whitehall II study. Alzheimer’s Res Ther 2021;13:1. https://doi.org/10.1186/s13195-020-00740-0.Search in Google Scholar PubMed PubMed Central

32. Le Guen, Y, Belloy, ME, Grenier-Boley, B, De Rojas, I, Castillo-Morales, A, Jansen, I, et al.. Association of rare APOE missense variants V236E and R251G with risk of Alzheimer disease. JAMA Neurol 2022;79:652–63. https://doi.org/10.1001/jamaneurol.2022.1166.Search in Google Scholar PubMed PubMed Central

33. Sienski, G, Narayan, P, Bonner, JM, Kory, N, Boland, S, Arczewska, AA, et al.. APOE4 disrupts intracellular lipid homeostasis in human iPSC-derived glia. Sci Transl Med 2021;13:eaaz4564. https://doi.org/10.1126/scitranslmed.aaz4564.Search in Google Scholar PubMed PubMed Central

34. Farmer, BC, Kluemper, J, Johnson, LA. Apolipoprotein E4 alters astrocyte fatty acid metabolism and lipid droplet formation. Cells 2019;8:182. https://doi.org/10.3390/cells8020182.Search in Google Scholar PubMed PubMed Central

35. Conte, M, Medici, V, Malagoli, D, Chiariello, A, Cirrincione, A, Davin, A, et al.. Expression pattern of perilipins in human brain during aging and in Alzheimer’s disease. Neuropathol Appl Neurobiol 2022;48:e12756. https://doi.org/10.1111/nan.12756.Search in Google Scholar PubMed PubMed Central

36. Sleegers, K, Bettens, K, De Roeck, A, Van Cauwenberghe, C, Cuyvers, E, Verheijen, J, et al.. A 22-single nucleotide polymorphism Alzheimer’s disease risk score correlates with family history, onset age, and cerebrospinal fluid Aβ42. Alzheimers Dement 2015;11:1452–60. https://doi.org/10.1016/j.jalz.2015.02.013.Search in Google Scholar PubMed

37. Sideris, DI, Danial, JSH, Emin, D, Ruggeri, FS, Xia, Z, Zhang, YP, et al.. Soluble amyloid beta-containing aggregates are present throughout the brain at early stages of Alzheimer’s disease. Brain Commun 2021;3:fcab147. https://doi.org/10.1093/braincomms/fcab147.Search in Google Scholar PubMed PubMed Central

38. Esparza, TJ, Wildburger, NC, Jiang, H, Gangolli, M, Cairns, NJ, Bateman, RJ, et al.. Soluble amyloid-beta aggregates from human Alzheimer’s disease brains. Sci Rep 2016;6:38187. https://doi.org/10.1038/srep38187.Search in Google Scholar PubMed PubMed Central

39. De, S, Wirthensohn, DC, Flagmeier, P, Hughes, C, Aprile, FA, Ruggeri, FS, et al.. Different soluble aggregates of Aβ42 can give rise to cellular toxicity through different mechanisms. Nat Commun 2019;10:1541. https://doi.org/10.1038/s41467-019-09477-3.Search in Google Scholar PubMed PubMed Central

40. Nag, S, Sarkar, B, Bandyopadhyay, A, Sahoo, B, Sreenivasan, VKA, Kombrabail, M, et al.. Nature of the amyloid-β monomer and the monomer-oligomer equilibrium. J Biol Chem 2011;286:13827–33. https://doi.org/10.1074/jbc.m110.199885.Search in Google Scholar PubMed PubMed Central

41. Hampel, H, Hardy, J, Blennow, K, Chen, C, Perry, G, Kim, SH, et al.. The amyloid-β pathway in Alzheimer’s disease. Mol Psychiatry 2021;26:5481–503. https://doi.org/10.1038/s41380-021-01249-0.Search in Google Scholar PubMed PubMed Central

42. Miura, T, Suzuki, K, Kohata, N, Takeuchi, H. Metal binding modes of Alzheimer’s amyloid β-peptide in insoluble aggregates and soluble complexes. Biochem 2000;39:7024–31. https://doi.org/10.1021/bi0002479.Search in Google Scholar PubMed

43. Razzokov, J, Yusupov, M, Bogaerts, A. Oxidation destabilizes toxic amyloid beta peptide aggregation. Sci Rep 2019;9:5476. https://doi.org/10.1038/s41598-019-41931-6.Search in Google Scholar PubMed PubMed Central

44. Ghosh, U, Thurber, KR, Yau, WM, Tycko, R. Molecular structure of a prevalent amyloid-β fibril polymorph from Alzheimer’s disease brain tissue. Proc Natl Acad Sci USA 2021;118:e2023089118. https://doi.org/10.1073/pnas.2023089118.Search in Google Scholar PubMed PubMed Central

45. Lee, M, Yau, WM, Louis, JM, Tycko, R. Structures of brain-derived 42-residue amyloid-β fibril polymorphs with unusual molecular conformations and intermolecular interactions. Proc Natl Acad Sci USA 2023;120:e2218831120. https://doi.org/10.1073/pnas.2218831120.Search in Google Scholar PubMed PubMed Central

46. Garbuz, DG, Zatsepina, OG, Evgen’ev, MB. Beta amyloid, tau protein, and neuroinflammation: an attempt to integrate different hypotheses of Alzheimer’s disease pathogenesis. Mol Biol 2021;55:670–82. https://doi.org/10.1134/s002689332104004x.Search in Google Scholar

47. Carrero, I, Gonzalo, MR, Martin, B, Sanz-Anquela, JM, Arevalo-Serrano, J, Gonzalo-Ruiz, A. Oligomers of beta-amyloid protein (Aβ1-42) induce the activation of cyclooxygenase-2 in astrocytes via an interaction with interleukin-1beta, tumour necrosis factor-alpha, and a nuclear factor kappa-B mechanism in the rat brain. Exp Neurol 2012;236:215–27. https://doi.org/10.1016/j.expneurol.2012.05.004.Search in Google Scholar PubMed

48. Chen, ZL, Singh, PK, Calvano, M, Norris, EH, Strickland, S. A possible mechanism for the enhanced toxicity of beta-amyloid protofibrils in Alzheimer’s disease. Proc Natl Acad Sci USA 2023;120:e2309389120. https://doi.org/10.1073/pnas.2309389120.Search in Google Scholar PubMed PubMed Central

49. Shea, D, Hsu, CC, Bi, TM, Paranjapye, N, Childers, MC, Cochran, J, et al.. α-Sheet secondary structure in amyloid β-peptide drives aggregation and toxicity in Alzheimer’s disease. Proc Natl Acad Sci USA 2019;116:8895–900. https://doi.org/10.1073/pnas.1820585116.Search in Google Scholar PubMed PubMed Central

50. Huang, HC, Tang, D, Xu, K, Jiang, ZF. Curcumin attenuates amyloid-β-induced tau hyperphosphorylation in human neuroblastoma SH-SY5Y cells involving PTEN/Akt/GSK-3β signaling pathway. J Recept Signal Transduct 2014;34:26–37. https://doi.org/10.3109/10799893.2013.848891.Search in Google Scholar PubMed

51. Basheer, N, Smolek, T, Hassan, I, Liu, F, Iqbal, K, Zilka, N, et al.. Does modulation of tau hyperphosphorylation represent a reasonable therapeutic strategy for Alzheimer’s disease? From preclinical studies to the clinical trials. Mol Psychiatry 2023;28:2197–214. https://doi.org/10.1038/s41380-023-02113-z.Search in Google Scholar PubMed PubMed Central

52. Wang, JZ, Xia, YY, Grundke-Iqbal, I, Iqbal, K. Abnormal hyperphosphorylation of tau: sites, regulation, and molecular mechanism of neurofibrillary degeneration. J Alzheimers Dis 2013;33:S123–39. https://doi.org/10.3233/jad-2012-129031.Search in Google Scholar

53. Tripathi, T, Khan, H. Direct interaction between the β-amyloid core and tau facilitates cross-seeding: a novel target for therapeutic intervention. Biochemistry 2020;59:341–2. https://doi.org/10.1021/acs.biochem.9b01087.Search in Google Scholar PubMed

54. Wölfl-Duchek, M, Mairinger, S, Hernández-Lozano, I, Filip, T, Zoufal, V, Löbsch, M, et al.. Use of PET imaging to assess the efficacy of thiethylperazine to stimulate cerebral MRP1 transport activity in wild-type and APP/PS1-21 mice. Int J Mol Sci 2022;23:6514. https://doi.org/10.3390/ijms23126514.Search in Google Scholar PubMed PubMed Central

55. North, WC, COLLAWN, TH, Hudnell, ABJr, Stephen, CR. Postoperative vomiting: influence of thiethylperazine. Anesth Analg 1963;42:559–65. https://doi.org/10.1213/00000539-196309000-00012.Search in Google Scholar

56. Weibel, S, Rücker, G, Eberhart, LH, Pace, NL, Hartl, HM, Jordan, OL, et al.. Drugs for preventing postoperative nausea and vomiting in adults after general anaesthesia: a network meta-analysis. Cochrane Database Syst Rev 2020;10:CD012859. https://doi.org/10.1002/14651858.cd012859.pub2.Search in Google Scholar

57. Scarpa, M, Molloy, C, Jenkins, L, Strellis, B, Budgett, RF, Hesse, S, et al.. Biased M1 muscarinic receptor mutant mice show accelerated progression of prion neurodegenerative disease. Proc Natl Acad Sci USA 2021;118:e2107389118. https://doi.org/10.1073/pnas.2107389118.Search in Google Scholar PubMed PubMed Central

58. Sheardown, MJ. Muscarinic M1 receptor agonists and M2 receptor antagonists as therapeutic targets in Alzheimer’s disease. Expert Opin Ther Pat 2002;12:863–70. https://doi.org/10.1517/13543776.12.6.863.Search in Google Scholar

59. Seeger, T, Fedorova, I, Zheng, F, Miyakawa, T, Koustova, E, Gomeza, J, et al.. M2 muscarinic acetylcholine receptor knock-out mice show deficits in behavioral flexibility, working memory, and hippocampal plasticity. J Neurosci 2004;24:10117–27. https://doi.org/10.1523/jneurosci.3581-04.2004.Search in Google Scholar

60. Darby, M, Schnoeller, C, Vira, A, Culley, F, Bobat, S, Logan, E, et al.. The M3 muscarinic receptor is required for optimal adaptive immunity to helminth and bacterial infection. PLoS Pathog 2015;11:e1004636. https://doi.org/10.1371/journal.ppat.1004636.Search in Google Scholar PubMed PubMed Central

61. Marsh, SE, Abud, EM, Lakatos, A, Karimzadeh, A, Yeung, ST, Davtyan, H, et al.. The adaptive immune system restrains Alzheimer’s disease pathogenesis by modulating microglial function. Proc Natl Acad Sci USA 2016;113:E1316–25. https://doi.org/10.1073/pnas.1525466113.Search in Google Scholar PubMed PubMed Central

62. Colloby, SJ, Nathan, PJ, McKeith, IG, Bakker, G, O’Brien, JT, Taylor, JP. Cholinergic muscarinic M1/M4 receptor networks in dementia with Lewy bodies. Brain Commun 2020;2:fcaa098. https://doi.org/10.1093/braincomms/fcaa098.Search in Google Scholar PubMed PubMed Central

63. Foster, DJ, Choi, DL, Conn, PJ, Rook, JM. Activation of M1 and M4 muscarinic receptors as potential treatments for Alzheimer’s disease and schizophrenia. Neuropsychiatr Dis Treat 2014:183–91. https://doi.org/10.2147/ndt.s55104.Search in Google Scholar PubMed PubMed Central

64. Araya, R, Noguchi, T, Yuhki, M, Kitamura, N, Higuchi, M, Saido, TC, et al.. Loss of M5 muscarinic acetylcholine receptors leads to cerebrovascular and neuronal abnormalities and cognitive deficits in mice. Neurobiol Dis 2006;24:334–44. https://doi.org/10.1016/j.nbd.2006.07.010.Search in Google Scholar PubMed

65. Perez, DM. α1-Adrenergic receptors: insights into potential therapeutic opportunities for COVID-19, heart failure, and Alzheimer’s disease. Int J Mol Sci 2023;24:4188. https://doi.org/10.3390/ijms24044188.Search in Google Scholar PubMed PubMed Central

66. Karczewski, P, Pohlmann, A, Wagenhaus, B, Wisbrun, N, Hempel, P, Lemke, B, et al.. Antibodies to the α1-adrenergic receptor cause vascular impairments in rat brain as demonstrated by magnetic resonance angiography. PLoS One;7:e41602. https://doi.org/10.1371/journal.pone.0041602.Search in Google Scholar PubMed PubMed Central

67. Iadecola, C, Duering, M, Hachinski, V, Joutel, A, Pendlebury, ST, Schneider, JA, et al.. Vascular cognitive impairment and dementia: JACC scientific expert panel. J Am Coll Cardiol 2019;73:3326–44. https://doi.org/10.1016/j.jacc.2019.04.034.Search in Google Scholar PubMed PubMed Central

68. Perez, DM. Current developments on the role of α1-adrenergic receptors in cognition, cardioprotection, and metabolism. Front Cell Dev Biol 2021;9:652152. https://doi.org/10.3389/fcell.2021.652152.Search in Google Scholar PubMed PubMed Central

69. Hao, L, Yang, Z, Lei, J. Underlying mechanisms of cooperativity, input specificity, and associativity of long-term potentiation through a positive feedback of local protein synthesis. Front Comput Neurosci 2018;12:25. https://doi.org/10.3389/fncom.2018.00025.Search in Google Scholar PubMed PubMed Central

70. Higuchi, M, Yanai, K, Okamura, N, Meguro, K, Arai, H, Itoh, M, et al.. Histamine H1 receptors in patients with Alzheimer’s disease assessed by positron emission tomography. Neuroscience 2000;99:721–9. https://doi.org/10.1016/s0306-4522(00)00230-x.Search in Google Scholar PubMed

71. Kárpáti, A, Yoshikawa, T, Naganuma, F, Matsuzawa, T, Kitano, H, Yamada, Y, et al.. Histamine H1 receptor on astrocytes and neurons controls distinct aspects of mouse behaviour. Sci Rep 2019;9:16451. https://doi.org/10.1038/s41598-019-52623-6.Search in Google Scholar PubMed PubMed Central

72. Yang, CC, Chien, WC, Chung, CH, Lai, CY, Tzeng, NS. The usage of histamine type 1 receptor antagonist and risk of dementia in the elderly: a nationwide cohort study. Front Aging Neurosci 2022;14:811494. https://doi.org/10.3389/fnagi.2022.811494.Search in Google Scholar PubMed PubMed Central

73. Pan, X, Kaminga, AC, Wen, SW, Wu, X, Acheampong, K, Liu, A. Dopamine and dopamine receptors in Alzheimer’s disease: a systematic review and network meta-analysis. Front Aging Neurosci 2019;11:175. https://doi.org/10.3389/fnagi.2019.00175.Search in Google Scholar PubMed PubMed Central

74. Francis, PT, Palmer, AM, Snape, M, Wilcock, GK. The cholinergic hypothesis of Alzheimer’s disease: a review of progress. J Neurol Neurosurg Psychiatry 1999;66:137–47. https://doi.org/10.1136/jnnp.66.2.137.Search in Google Scholar PubMed PubMed Central

75. Zhang, G, Stackman, RWJr. The role of serotonin 5-HT2A receptors in memory and cognition. Front Pharmacol 2015;6:225. https://doi.org/10.3389/fphar.2015.00225.Search in Google Scholar PubMed PubMed Central

76. Celada, P, Puig, MV, Amargós-Bosch, M, Adell, A, Artigas, F. The therapeutic role of 5-HT1A and 5-HT2A receptors in depression. J Psychiatry Neurosci 2004;29:252–65.Search in Google Scholar

77. Tekin, S, Cummings, JL. Frontal–subcortical neuronal circuits and clinical neuropsychiatry: an update. J Psychosom Res 2002;53:647–54. https://doi.org/10.1016/s0022-3999(02)00428-2.Search in Google Scholar PubMed

78. Baudic, S, Dalla Barba, G, Thibaudet, MC, Smagghe, A, Remy, P, Traykov, L. Executive function deficits in early Alzheimer’s disease and their relations with episodic memory. Arch Clin Neuropsychol 2006;21:15–21. https://doi.org/10.1016/j.acn.2005.07.002.Search in Google Scholar PubMed

79. Riveros, R, Bakchine, S, Pillon, B, Poupon, F, Miranda, M, Slachevsky, A. Fronto-subcortical circuits for cognition and motivation: dissociated recovery in a case of loss of psychic self-activation. Front Psychol 2019;9:2781. https://doi.org/10.3389/fpsyg.2018.02781.Search in Google Scholar PubMed PubMed Central

80. Millan, MJ. Serotonin 5-HT2C receptors as a target for the treatment of depressive and anxious states: focus on novel therapeutic strategies. Therapies 2005;60:441–60. https://doi.org/10.2515/therapie:2005065.10.2515/therapie:2005065Search in Google Scholar PubMed

81. Reeves, S, Brown, R, Howard, R, Grasby, P. Increased striatal dopamine (D2/D3) receptor availability and delusions in Alzheimer disease. Neurology 2009;72:528–34. https://doi.org/10.1212/01.wnl.0000341932.21961.f3.Search in Google Scholar PubMed PubMed Central

82. Ismail, Z, Creese, B, Aarsland, D, Kales, HC, Lyketsos, CG, Sweet, RA, et al.. Psychosis in Alzheimer disease – mechanisms, genetics and therapeutic opportunities. Nat Rev Neurol 2022;18:131–44. https://doi.org/10.1038/s41582-021-00597-3.Search in Google Scholar PubMed PubMed Central

83. Ropacki, SA, Jeste, DV. Epidemiology of and risk factors for psychosis of Alzheimer’s disease: a review of 55 studies published from 1990 to 2003. Am J Psychiatry 2005;162:2022–30. https://doi.org/10.1176/appi.ajp.162.11.2022.Search in Google Scholar PubMed

84. Holmes, C, Arranz, M, Collier, D, Powell, J, Lovestone, S. Depression in Alzheimer’s disease: the effect of serotonin receptor gene variation. Am J Med Genet B: Neuropsychiatr Genet 2003;119:40–3. https://doi.org/10.1002/ajmg.b.10068.Search in Google Scholar PubMed

85. Izzo, NJ, Yuede, CM, LaBarbera, KM, Limegrover, CS, Rehak, C, Yurko, R, et al.. Preclinical and clinical biomarker studies of CT1812: a novel approach to Alzheimer’s disease modification. Alzheimers Dement 2021;17:1365–82. https://doi.org/10.1002/alz.12302.Search in Google Scholar PubMed PubMed Central

86. Li, Y, Rusinek, H, Butler, T, Glodzik, L, Pirraglia, E, Babich, J, et al.. Decreased CSF clearance and increased brain amyloid in Alzheimer’s disease. Fluids Barriers CNS 2022;19:1–9. https://doi.org/10.1186/s12987-022-00318-y.Search in Google Scholar PubMed PubMed Central

87. Yang, T, Li, S, Xu, H, Walsh, DM, Selkoe, DJ. Large soluble oligomers of amyloid β-protein from Alzheimer brain are far less neuroactive than the smaller oligomers to which they dissociate. J Neurosci 2017;37:152–63. https://doi.org/10.1523/jneurosci.1698-16.2017.Search in Google Scholar

88. Silverberg, GD, Mayo, M, Saul, T, Rubenstein, E, McGuire, D. Alzheimer’s disease, normal-pressure hydrocephalus, and senescent changes in CSF circulatory physiology: a hypothesis. Lancet Neurol 2003;2:506–11. https://doi.org/10.1016/s1474-4422(03)00487-3.Search in Google Scholar PubMed

89. Kang, MJ, Park, YH, Kim, S, Yi, S. Hydrocephalus in a patient with Alzheimer’s disease. Dement Neurocogn Disord 2018;17:32–6. https://doi.org/10.12779/dnd.2018.17.1.32.Search in Google Scholar PubMed PubMed Central

90. Hölttä, M, Hansson, O, Andreasson, U, Hertze, J, Minthon, L, Nägga, K, et al.. Evaluating amyloid-β oligomers in cerebrospinal fluid as a biomarker for Alzheimer’s disease. PLoS One 2013;8:e66381. https://doi.org/10.1371/journal.pone.0066381.Search in Google Scholar PubMed PubMed Central

91. Fukumoto, H, Tokuda, T, Kasai, T, Ishigami, N, Hidaka, H, Kondo, M, et al.. High-molecular-weight β-amyloid oligomers are elevated in cerebrospinal fluid of Alzheimer patients. FASEB J 2010;24:2716–26. https://doi.org/10.1096/fj.09-150359.Search in Google Scholar PubMed

92. Yasumoto, T, Takamura, Y, Tsuji, M, Watanabe-Nakayama, T, Imamura, K, Inoue, H, et al.. High molecular weight amyloid β1-42 oligomers induce neurotoxicity via plasma membrane damage. FASEB J 2019;33:9220–34. https://doi.org/10.1096/fj.201900604r.Search in Google Scholar PubMed

93. Huang, YR, Liu, RT. The toxicity and polymorphism of β-amyloid oligomers. Int J Mol Sci 2020;21:4477. https://doi.org/10.3390/ijms21124477.Search in Google Scholar PubMed PubMed Central

94. Meilandt, WJ, Maloney, JA, Imperio, J, Lalehzadeh, G, Earr, T, Crowell, S, et al.. Characterization of the selective in vitro and in vivo binding properties of crenezumab to oligomeric Aβ. Alzheimer’s Res Ther 2019;11:1–4. https://doi.org/10.1186/s13195-019-0553-5.Search in Google Scholar PubMed PubMed Central

95. Apátiga-Pérez, R, Soto-Rojas, LO, Campa-Córdoba, BB, Luna-Viramontes, NI, Cuevas, E, Villanueva-Fierro, I, et al.. Neurovascular dysfunction and vascular amyloid accumulation as early events in Alzheimer’s disease. Metab Brain Dis 2022;37:39–50. https://doi.org/10.1007/s11011-021-00814-4.Search in Google Scholar PubMed

96. Smith, EE, Greenberg, SM. β-Amyloid, blood vessels, and brain function. Stroke 2009;40:2601–6. https://doi.org/10.1161/strokeaha.108.536839.Search in Google Scholar

97. Greenberg, SM, Gurol, ME, Rosand, J, Smith, EE. Amyloid angiopathy – related vascular cognitive impairment. Stroke 2004;35:2616–9. https://doi.org/10.1161/01.str.0000143224.36527.44.Search in Google Scholar

98. Fuller, JP, Stavenhagen, JB, Christensen, S, Kartberg, F, Glennie, MJ, Teeling, JL. Comparing the efficacy and neuroinflammatory potential of three anti-abeta antibodies. Acta Neuropathol 2015;130:699–711. https://doi.org/10.1007/s00401-015-1484-2.Search in Google Scholar PubMed PubMed Central

99. Guthrie, H, Honig, LS, Lin, H, Sink, KM, Blondeau, K, Quartino, A, et al.. Safety, tolerability, and pharmacokinetics of crenezumab in patients with mild-to-moderate Alzheimer’s disease treated with escalating doses for up to 133 weeks. J Alzheimers Dis 2020;76:967–79. https://doi.org/10.3233/jad-200134.Search in Google Scholar PubMed PubMed Central

100. Yang, T, Dang, Y, Ostaszewski, B, Mengel, D, Steffen, V, Rabe, C, et al.. Target engagement in an Alzheimer trial: crenezumab lowers amyloid β oligomers in cerebrospinal fluid. Ann Neurol 2019;86:215–24. https://doi.org/10.1002/ana.25513.Search in Google Scholar PubMed PubMed Central

101. Kravenska, Y, Nieznanska, H, Nieznanski, K, Lukyanetz, E, Szewczyk, A, Koprowski, P. The monomers, oligomers, and fibrils of amyloid-β inhibit the activity of mitoBKCa channels by a membrane-mediated mechanism. Biochim Biophys Acta Biomembr 2020;1862:183337. https://doi.org/10.1016/j.bbamem.2020.183337.Search in Google Scholar PubMed

102. Neumann, U, Ufer, M, Jacobson, LH, Rouzade-Dominguez, ML, Huledal, G, Kolly, C, et al.. The BACE-1 inhibitor CNP 520 for prevention trials in Alzheimer’s disease. EMBO Mol Med 2018;10:e9316. https://doi.org/10.15252/emmm.201809316.Search in Google Scholar PubMed PubMed Central

103. Yan, R. Physiological functions of the β-site amyloid precursor protein cleaving enzyme 1 and 2. Front Mol Neurosci 2017;10:97. https://doi.org/10.3389/fnmol.2017.00097.Search in Google Scholar PubMed PubMed Central

104. Hu, X, He, W, Luo, X, Tsubota, KE, Yan, R. BACE1 regulates hippocampal astrogenesis via the Jagged1-Notch pathway. Cell Rep 2013;4:40–9. https://doi.org/10.1016/j.celrep.2013.06.005.Search in Google Scholar PubMed PubMed Central

105. Geoffroy, CG, Meves, JM, Zheng, B. The age factor in axonal repair after spinal cord injury: a focus on neuron-intrinsic mechanisms. Neurosci Lett 2017;652:41–9. https://doi.org/10.1016/j.neulet.2016.11.003.Search in Google Scholar PubMed PubMed Central

106. Jaerve, A, Schiwy, N, Schmitz, C, Mueller, HW. Differential effect of aging on axon sprouting and regenerative growth in spinal cord injury. Exp Neurol 2011;231:284–94. https://doi.org/10.1016/j.expneurol.2011.07.002.Search in Google Scholar PubMed

107. Bradke, F. Mechanisms of axon growth and regeneration: moving between development and disease. J Neurosci 2022;42:8393–405. https://doi.org/10.1523/jneurosci.1131-22.2022.Search in Google Scholar

108. Chan, WM, Traboulsi, EI, Arthur, B, Friedman, N, Andrews, C, Engle, EC. Horizontal gaze palsy with progressive scoliosis can result from compound heterozygous mutations in ROBO3. J Med Genet 2006;43:e11. https://doi.org/10.1136/jmg.2005.035436.Search in Google Scholar PubMed PubMed Central

109. Nugent, AA, Kolpak, AL, Engle, EC. Human disorders of axon guidance. Curr Opin Neurobiol 2012;22:837–43. https://doi.org/10.1016/j.conb.2012.02.006.Search in Google Scholar PubMed PubMed Central

110. Stampanoni Bassi, M, Iezzi, E, Gilio, L, Centonze, D, Buttari, F. Synaptic plasticity shapes brain connectivity: implications for network topology. Int J Mol Sci 2019;20:6193. https://doi.org/10.3390/ijms20246193.Search in Google Scholar PubMed PubMed Central

111. Van Dyck, CH, Swanson, CJ, Aisen, P, Bateman, RJ, Chen, C, Gee, M, et al.. Lecanemab in early Alzheimer’s disease. N Engl J Med 2023;388:9–21. https://doi.org/10.1056/nejmoa2212948.Search in Google Scholar PubMed

112. Siddiqi, MK, Majid, N, Malik, S, Alam, P, Khan, RH. Amyloid oligomers, protofibrils and fibrils. Subcell Biochem 2019;93:471–503. https://doi.org/10.1007/978-3-030-28151-9_16.Search in Google Scholar PubMed

113. Ono, K, Tsuji, M. Protofibrils of amyloid-β are important targets of a disease-modifying approach for Alzheimer’s disease. Int J Mol Sci 2020;21:952. https://doi.org/10.3390/ijms21030952.Search in Google Scholar PubMed PubMed Central

114. Tedoldi, A, Ludwig, P, Fulgenzi, G, Takeshima, H, Pedarzani, P, Stocker, M. Calcium-induced calcium release and type 3 ryanodine receptors modulate the slow after hyperpolarising current, sIAHP, and its potentiation in hippocampal pyramidal neurons. PLoS One 2020;15:e0230465. https://doi.org/10.1371/journal.pone.0230465.Search in Google Scholar PubMed PubMed Central

115. Tong, BC, Wu, AJ, Li, M, Cheung, KH. Calcium signaling in Alzheimer’s disease & therapies. Biochim Biophys Acta Mol Cell Res 2018;1865:1745–60. https://doi.org/10.1016/j.bbamcr.2018.07.018.Search in Google Scholar PubMed

116. Baracaldo-Santamaría, D, Avendaño-Lopez, SS, Ariza-Salamanca, DF, Rodriguez-Giraldo, M, Calderon-Ospina, CA, González-Reyes, RE, et al.. Role of calcium modulation in the pathophysiology and treatment of Alzheimer’s disease. Int J Mol Sci 2023;24:9067. https://doi.org/10.3390/ijms24109067.Search in Google Scholar PubMed PubMed Central

117. Oulès, B, Del Prete, D, Greco, B, Zhang, X, Lauritzen, I, Sevalle, J, et al.. Ryanodine receptor blockade reduces amyloid-β load and memory impairments in Tg2576 mouse model of Alzheimer disease. J Neurosci 2012;32:11820–34. https://doi.org/10.1523/jneurosci.0875-12.2012.Search in Google Scholar PubMed PubMed Central

118. Williams, AD, Sega, M, Chen, M, Kheterpal, I, Geva, M, Berthelier, V, et al.. Structural properties of Aβ protofibrils stabilized by a small molecule. Proc Natl Acad Sci USA 2005;102:7115–20. https://doi.org/10.1073/pnas.0408582102.Search in Google Scholar PubMed PubMed Central

119. An, J, Kim, K, Lim, HJ, Kim, HY, Shin, J, Park, I, et al.. Early onset diagnosis in Alzheimer’s disease patients via amyloid-β oligomers-sensing probe in cerebrospinal fluid. Nat Commun 2024;15:1004. https://doi.org/10.1038/s41467-024-44818-x.Search in Google Scholar PubMed PubMed Central

120. Govindpani, K, McNamara, LG, Smith, NR, Vinnakota, C, Waldvogel, HJ, Faull, RL, et al.. Vascular dysfunction in Alzheimer’s disease: a prelude to the pathological process or a consequence of it? J Clin Med 2019;8:651. https://doi.org/10.3390/jcm8050651.Search in Google Scholar PubMed PubMed Central

121. Passeri, E, Elkhoury, K, Morsink, M, Broersen, K, Linder, M, Tamayol, A, et al.. Alzheimer’s disease: treatment strategies and their limitations. Int J Mol Sci 2022;23:13954. https://doi.org/10.3390/ijms232213954.Search in Google Scholar PubMed PubMed Central

122. Ullah, SNMN, Afzal, O, Altamimi, ASA, Ather, H, Sultana, S, Almalki, WH, et al.. Nanomedicine in the management of Alzheimer’s disease: state-of-the-art. Biomedicines 2023;11:1752. https://doi.org/10.3390/biomedicines11061752.Search in Google Scholar PubMed PubMed Central

123. Giuffrida, ML, Caraci, F, Pignataro, B, Cataldo, S, De Bona, P, Bruno, V, et al.. β-amyloid monomers are neuroprotective. J Neurosci 2009;29:10582–7. https://doi.org/10.1523/jneurosci.1736-09.2009.Search in Google Scholar PubMed PubMed Central

124. Sun, D, Gao, W, Hu, H, Zhou, S. Why 90% of clinical drug development fails and how to improve it? Acta Pharm Sin B 2022;12:3049–62. https://doi.org/10.1016/j.apsb.2022.02.002.Search in Google Scholar PubMed PubMed Central

125. Varshavskaya, KB, Mitkevich, VA, Makarov, AA, Barykin, EP. Synthetic, cell-derived, brain-derived, and recombinant β-amyloid: modelling Alzheimer’s disease for research and drug development. Int J Mol Sci 2022;23:15036. https://doi.org/10.3390/ijms232315036.Search in Google Scholar PubMed PubMed Central

Received: 2023-12-29
Accepted: 2024-02-28
Published Online: 2024-03-18

© 2024 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 29.5.2024 from https://www.degruyter.com/document/doi/10.1515/jbcpp-2023-0264/html
Scroll to top button