Alzheimer's Disease in Clinical Trials
About Alzheimer’s Disease
Alzheimer’s disease is a progressive brain disorder that gradually destroys memory and thinking skills, eventually impairing the ability to perform everyday tasks. It is a specific type of dementia, which is a broader term describing symptoms that affect memory, thinking, and social abilities. Alzheimer’s is most common in older adults; however, it is not a normal part of aging. Instead, it is caused by complex changes in the brain that lead to the loss of brain cells.
One of the main causes of Alzheimer’s disease is the abnormal buildup of amyloid and tau proteins. In 1984, G. Glenner and C. Wong discovered that amyloid-beta (Aβ) proteins are the central component of the extracellular amyloid plaques found in Alzheimer’s disease (Glenner and Wong 1984). A year later, researchers identified tau protein as the key component forming neurofibrillary tangles, another hallmark of the disease (Grundke-Iqbal et al. 1986, Delacourte and Défossez 1986).
The timeline of research targeting amyloid-beta and tau proteins in Alzheimer’s disease (AD) reflects a shift in scientific focus. Aβ was identified in the 1980s, since then sparking decades of attempts to develop anti-Aβ therapies. While most early trials failed, the early 2020s saw the approval of Aβ-clearing antibodies such as lecanemab (Leqembi®), which received accelerated approval from the U.S. FDA in January 2023 and was later converted to traditional approval after the Phase 3 CLARITY AD trial verified clinical benefit (van Dyck et al., 2023; U.S. FDA, 2023), and aducanumab (Aduhelm®), approved by the FDA in June 2021 under the accelerated approval pathway based on its effect of reducing amyloid-beta plaques (U.S. FDA, 2021). More recently, attention has turned to tau pathology, which is closely linked to neurodegeneration. Multiple anti-tau therapies are now in clinical trials. Current strategies suggest that the most effective treatments may need to target both Aβ and tau together to achieve better outcomes.
How are Alzheimer’s Drugs Validated in IND-Enabling and Phase I Trials
In Alzheimer’s drug development, biomarkers such as amyloid and tau levels are used to confirm the presence of disease-related targets and to demonstrate a drug’s ability to interact with those targets. This process helps validate the drug’s mechanism of action in humans. In Phase I clinical trials, Alzheimer’s drugs are tested for safety and appropriate dosage in a small group of healthy volunteers. These studies evaluate the drug’s absorption, distribution, metabolism, excretion, and toxicity (ADMET). Before Phase I begins, an Investigational New Drug (IND) application must be submitted to the FDA. This regulatory process authorises first-in-human testing and helps determine the drug’s pharmacokinetics and maximum tolerated dose (MTD), which are critical for designing subsequent, larger trials. Phase I studies for Alzheimer’s drugs often use advanced methods and tools such as magnetic resonance imaging (MRI) and position emission tomography (PET) to track biomarkers like amyloid and tau, as well as computational approaches including artificial intelligence (AI) and digital sensors. These technologies not only assess drug safety, target engagement, and disease progression, but also generate more sophisticated data than traditional methods.
Electrophysiological Tools
Methods such as electroencephalography (EEG) and transcranial magnetic stimulation (TMS) are increasingly used to evaluate brain network activity, cortical excitability, and drug-induced changes in neural function (Maiella et al. 2024). These techniques provide sensitive, real-time measures of target engagement and can help refine dosing, improve patient stratification, and offer early signals of therapeutic efficacy.
Enhanced Clinical Trial Efficiency
One major development in enhancing clinical trial efficiency is the application of precision medicine approaches, where AI, advanced neuroimaging, and electrophysiological tools are employed to stratify patient populations more accurately. By identifying individuals most likely to benefit from a given therapy, these tools reduce heterogeneity in trial cohorts and increase the likelihood of detecting treatment effects, ultimately accelerating the validation of novel interventions, particularly in the attempts to advance novel Alzheimer’s interventions (Reitz 2016).
In parallel, the adoption of remote monitoring technologies has expanded the capacity to collect continuous, real-world data outside for traditional clinical visits. Wearable sensors, smartphone-based assessments, and digital biomarkers enable longitudinal tracking of symptoms, functional changes and disease progression with higher temporal resolution than conventional endpoints (Muurling et al. 2021). Such approaches not only improve patient compliance and engagement but also provide datasets that better reflect everyday experiences. Particularly important in the Alzheimer’s population where their day-to-day interactions and behaviours are most affected.
Mechanisms of Action for Drugs Treating Alzheimer’s
Alzheimer’s disease is increasingly understood as a disorder of the synaptic microenvironment. Beyond amyloid plaques and tau tangles, early pathophysiology occurs at the synapse, where an imbalance in neurotransmission destabilises neuronal networks. A critical player here is the NMDA receptor (NMDAR), which normally regulates calcium influx for synaptic plasticity and memory formation (Liu et al. 2019). In Alzheimer’s, excessive glutamate release, impaired astrocytic uptake, and altered receptor subunit composition drive NMDAR overactivation, leading to calcium overload, mitochondrial stress, and ultimately excitotoxicity. Simultaneously, amyloid-beta oligomers and tau pathology disrupt receptor trafficking, weaken dendritic spines, and impair long-term potentiation. The surrounding synaptic milieu (astrocytes, microglia, and extracellular matrix proteins) further contributes by releasing inflammatory cytokines, failing to buffer glutamate, or pruning synapses excessively. Current drugs like memantine partially restore balance by blocking pathological NMDAR activity while sparing physiological signalling (Kabir et al. 2019).
However, researchers are now exploring combinatorial strategies that target amyloid, tau, and synaptic dysfunction simultaneously to preserve network integrity (Maramai et al. 2020).
Future Directions for Clinical Trials in Alzheimer’s Populations
Future IND-enabling and early Phase I Alzheimer’s trials must move beyond single-target strategies toward combinational approaches that address amyloid, tau, neuroinflammation, and synaptic dysfunction simultaneously (Abbas et al. 2025). Incorporating AI for patient stratification, predictive modelling, and adaptive trial design will streamline development and reduce failure rates. Equally important is the integration of neurophysiological and electrophysiological tools to provide real-time biomarkers of target engagement and network health (Shaheen et al. 2025 and Rudroff et al. 2024). Together, these innovations will accelerate translation of experimental therapies, broaden treatment options, and bring more effective and personalised drugs to patients with Alzheimer’s disease.
References:
-
Abbas, K., Mustafa, M., Alam, M. et al., 2025. Multi-target approach to Alzheimer’s disease prevention and treatment: antioxidant, anti-inflammatory, and amyloid-modulating mechanisms. Neurogenetics, 26, p.39. https://doi.org/10.1007/s10048-025-00821-y
-
Delacourte, A. and Défossez, A., 1986. Alzheimer’s disease: Tau proteins, the promoting factors of microtubule assembly, are major components of paired helical filaments. Journal of the Neurological Sciences, 76(2–3), pp.173–186. https://doi.org/10.1016/0022-510X(86)90167-X
-
Glenner, G.G. and Wong, C.W., 1984. Alzheimer’s disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochemical and Biophysical Research Communications, 120(3), pp.885–890. https://doi.org/10.1016/S0006-291X(84)80190-4
-
Grundke-Iqbal, I., Iqbal, K., Quinlan, M., Tung, Y.C., Zaidi, M.S. and Wisniewski, H.M., 1986. Microtubule-associated protein tau. A component of Alzheimer paired helical filaments. Journal of Biological Chemistry, 261(13), pp.6084–6089. Available at: https://pubmed.ncbi.nlm.nih.gov/3084478/
-
Høilund-Carlsen, P.F., Alavi, A. and Barrio, J.R., 2024. PET/CT/MRI in clinical trials of Alzheimer’s disease. Journal of Alzheimer’s Disease, 101(s1), pp.S579–S601. https://doi.org/10.3233/JAD-240206
-
Kabir, M.T., Sufian, M.A., Uddin, M.S., Begum, M.M., Akhter, S., Islam, A., Mathew, B., Islam, M.S., Amran, M.S. and Ashraf, G.M., 2019. NMDA receptor antagonists: repositioning of memantine as a multitargeting agent for Alzheimer’s therapy. Current Pharmaceutical Design, 25(33), pp.3506–3518.
-
Liu, J., Chang, L., Song, Y., Li, H. and Wu, Y., 2019. The role of NMDA receptors in Alzheimer’s disease. Frontiers in Neuroscience, 13, p.43. https://doi.org/10.3389/fnins.2019.00043
-
Maiella, M., Mencarelli, L., Casula, E.P., Borghi, I., Assogna, M., di Lorenzo, F., Bonnì, S., Pezzopane, V., Martorana, A. and Koch, G., 2024. Breakdown of TMS evoked EEG signal propagation within the default mode network in Alzheimer’s disease. Clinical Neurophysiology, 167, pp.177–188. https://doi.org/10.1016/j.clinph.2024.09.007
-
Maramai, S., Benchekroun, M., Gabr, M.T. and Yahiaoui, S., 2020. Multitarget therapeutic strategies for Alzheimer’s disease: Review on emerging target combinations. BioMed Research International, 2020, Article ID 5120230.
-
Muurling, M., de Boer, C., Kozak, R. et al., 2021. Remote monitoring technologies in Alzheimer’s disease: design of the RADAR-AD study. Alzheimer’s Research & Therapy, 13, p.89. https://doi.org/10.1186/s13195-021-00825-4
-
Noda, K., Kageyama, I., Kobayashi, Y., Lim, Y., Sengoku, S. and Kodama, K., 2025. Leveraging mHealth wearables for managing patients with Alzheimer’s disease: a scoping review. Drug Discovery Today, 30(5), p.104363. https://doi.org/10.1016/j.drudis.2025.104363
-
Reitz, C., 2016. Toward precision medicine in Alzheimer’s disease. Annals of Translational Medicine, 4(6), p.107. https://doi.org/10.21037/atm.2016.03.05
-
Rudroff, T., Rainio, O. and Klén, R., 2024. AI for the prediction of early stages of Alzheimer’s disease from neuroimaging biomarkers – A narrative review of a growing field. Neurological Sciences, 45, pp.5117–5127. https://doi.org/10.1007/s10072-024-07649-8
-
Shaheen, H. and Melnik, R., 2025. Brain network dynamics and multiscale modelling of neurodegenerative disorders: a review. IEEE Access, 13, pp.33074–33100. https://doi.org/10.1109/ACCESS.2025.3542634
-
U.S. Food and Drug Administration, 2021. FDA’s decision to approve new treatment for Alzheimer’s disease (Aduhelm). 7 June. Available at: https://www.fda.gov/drugs/our-perspective/fdas-decision-approve-new-treatment-alzheimers-disease
-
U.S. Food and Drug Administration, 2023. FDA converts novel Alzheimer’s disease treatment to traditional approval — Leqembi (lecanemab-irmb). 6 July. Available at: https://www.fda.gov/news-events/press-announcements/fda-converts-novel-alzheimers-disease-treatment-traditional-approval
-
van Dyck, C.H. et al., 2023. Lecanemab in early Alzheimer’s disease. New England Journal of Medicine, 389(1), pp.9–21. https://doi.org/10.1056/NEJMoa2212948