View Article

  • Pharmacological Approaches in Alzheimer’s Disease: From Symptomatic Care to Future Disease Modification

  • Koringa College of Pharmacy, Korangi, Kakinada, Andhra Pradesh

Abstract

Alzheimer’s disease (AD) is the most prevalent cause of dementia worldwide, posing immense medical, social, and economic challenges. Despite decades of research, pharmacological management has largely remained limited to symptomatic treatments, with cholinesterase inhibitors and NMDA receptor antagonists providing modest benefit. However, recent advances in understanding AD pathophysiology—particularly amyloid-? aggregation, tau pathology, neuroinflammation, and vascular contributions—have led to the development of disease-modifying therapies (DMTs). Monoclonal antibodies such as aducanumab, lecanemab, and donanemab have shown potential to slow cognitive decline, though challenges of safety, cost, and accessibility persist. Parallel research into anti-tau strategies, immunotherapies, multi-target drugs, and precision medicine approaches holds further promise. In addition to pharmacological interventions, non-pharmacological strategies—including lifestyle modifications, dietary interventions, physical activity, cognitive training, and psychosocial support—remain essential for holistic management of AD. Looking forward, advances in biomarker-guided therapy, stem cell research, gene editing, and artificial intelligence are shaping the future landscape of AD treatment. While curative therapies remain elusive, the convergence of pharmacological innovation with preventive and supportive strategies offers renewed hope for patients and caregivers. This review synthesizes current knowledge, highlights recent pharmacological advances, and explores future directions in the management of Alzheimer’s disease.

Keywords

Alzheimer’s disease; pharmacology; disease-modifying therapies; amyloid; tau; biomarkers; neuroinflammation; precision medicine; stem cells; non-pharmacological interventions; dementia management.

Introduction

Alzheimer’s disease (AD) has emerged as one of the most pressing neurological disorders of the 21st century. First described in 1906 by Alois Alzheimer in a patient with profound memory loss, emotional instability, and neuropathological hallmarks of amyloid plaques and neurofibrillary tangles, the disease has since become a global health challenge. Currently, over 55 million individuals are living with dementia, of which 60–70% are attributable to AD¹. This number is projected to triple by 2050 due to aging populations, particularly in low- and middle-income countries².The burden of AD extends beyond patients to families, caregivers, and health systems. Annual global costs are estimated to exceed USD 1 trillion, with both direct medical expenses and indirect costs such as lost productivity and caregiver stress³. Importantly, AD is not merely a neurobiological condition but also a deeply human disease that strips individuals of memory, identity, and independence, creating emotional suffering for patients and loved ones.Pharmacological interventions, although essential, remain limited in their ability to halt or reverse disease progression. Current therapies primarily offer symptomatic relief, modestly improving cognition and daily functioning. However, recent scientific breakthroughs — including monoclonal antibodies targeting amyloid-β — represent a paradigm shift toward disease-modifying treatments?. Despite controversies, these innovations signal a hopeful transition from palliative symptom control to potentially altering the disease trajectory.This review aims to provide a comprehensive, detailed exploration of the pharmacological landscape of AD. It integrates historical context, molecular insights, current and emerging drug therapies, adjunctive approaches, and future directions. In doing so, it highlights not only scientific progress but also the urgent need for compassionate, patient-centered strategies that preserve dignity and quality of life.

2. Historical Background of Alzheimer’s Disease

The story of Alzheimer’s disease began in 1906, when Alois Alzheimer presented the case of Auguste Deter, a 51-year-old woman with progressive memory loss, paranoia, and cognitive decline. Upon her death, postmortem examination revealed hallmark neuropathological features — extracellular “senile plaques” and intracellular “neurofibrillary tangles.” These observations laid the foundation for identifying AD as a distinct neurodegenerative disorder¹.

Over the decades, advances in neuropathology, biochemistry, and genetics deepened understanding of the disease. In the 1970s, researchers highlighted the cholinergic deficit hypothesis, noting significant reductions in acetylcholine levels in AD patients. This led to the development of cholinesterase inhibitors, the first class of FDA-approved drugs for AD². The 1980s and 1990s brought two landmark discoveries: the amyloid cascade hypothesis, which proposed amyloid-β accumulation as a key driver of pathology, and the tau hypothesis, which focused on abnormal tau hyperphosphorylation and tangle formation³.

The late 1990s and early 2000s witnessed approval of memantine, an NMDA receptor antagonist, as well as large-scale clinical trials targeting amyloid-β. Despite repeated trial failures, these efforts shaped the modern research landscape. By the 2010s, genetic insights — particularly the role of the APOE ε4 allele — underscored the interplay between genes and environment?.

The 2020s marked a turning point with the controversial approval of aducanumab in 2021, followed by promising results for lecanemab and donanemab. For the first time, therapies aimed not just to relieve symptoms but to modify the underlying disease process?. Though debates continue regarding efficacy, safety, and affordability, these approvals represent historic milestones in the century-long fight against AD.

Table 1: Key Historical Milestones in Alzheimer’s Disease Research

Year

Milestone

Impact

1906

Alois Alzheimer describes plaques & tangles in Auguste Deter

First identification of AD pathology

1970s

Cholinergic hypothesis proposed

Led to development of cholinesterase inhibitors

1984

Amyloid-β protein identified

Foundation of amyloid cascade hypothesis

1986

Tau protein abnormalities described

Birth of tau hypothesis

1993

APOE ε4 identified as genetic risk factor

Opened doors to pharmacogenomics research

1996–2003

Donepezil, rivastigmine, galantamine, and memantine approved

First pharmacological treatments for AD

2021

Aducanumab FDA approval

First disease-modifying therapy (controversial)

2023

Lecanemab& donanemab trials show positive outcomes

Renewed hope for disease modification

3. Epidemiology, Burden, and Risk Factors

Alzheimer’s disease (AD) is the leading cause of dementia, accounting for 60–70% of cases worldwide¹. According to the World Health Organization, more than 55 million people live with dementia in 2023, a number projected to rise to 139 million by 2050 due to aging populations². This makes AD not only a medical challenge but also a global public health crisis.

3.1 Global Prevalence

Prevalence rates vary significantly across regions. In North America and Europe, where life expectancy is higher, prevalence of AD in individuals above 65 years ranges from 5–8%. In Asia, rapid aging, especially in China and India, is expected to contribute to a majority of global dementia cases by mid-century³. Meanwhile, in Africa and low-income countries, prevalence data remain limited, but underdiagnosis is common due to poor healthcare infrastructure?.

3.2 Socioeconomic Burden

The economic burden of AD is staggering. The global cost of dementia was estimated at over USD 1 trillion in 2020 and is expected to double by 2030?. Direct medical costs include diagnosis, hospitalizations, and medications, while indirect costs include caregiver time, lost productivity, and social support. Importantly, up to 80% of dementia care is provided by unpaid family caregivers, leading to financial strain, emotional exhaustion, and health issues?.

Figure-1

3.3 Risk Factors

AD arises from a complex interplay of non-modifiable and modifiable risk factors.

  • Non-modifiable factors: Age remains the strongest risk factor. Genetics, particularly the APOE ε4 allele, increases risk up to 3-fold for heterozygotes and 12-fold for homozygotes?.
  • Modifiable factors: Cardiovascular health, diabetes, hypertension, obesity, smoking, and physical inactivity contribute significantly to risk. Increasing evidence also links diet, social isolation, depression, and chronic stress to AD onset?.
  • Protective factors: Higher education, cognitive reserve, active lifestyles, and Mediterranean-style diets appear to lower risk?.

Table 2: Global Prevalence and Burden of Alzheimer’s Disease

Region

Prevalence in ≥65 years

Estimated Patients (2023)

Projected Patients (2050)

Notes

North America

6–8%

~7 million

~14 million

High healthcare expenditure, advanced diagnostics

Europe

6–7%

~10 million

~18 million

Aging population, high prevalence in women

Asia

4–6%

~22 million

~70 million

Rapid demographic shift, underdiagnosis in rural areas

Africa

2–4%

~4 million

~10 million

Data gaps, limited resources

Global

~5–7%

~55 million (all dementias)

~139 million

Major economic and social burden

4. Pathophysiology of Alzheimer’s Disease

Alzheimer’s disease (AD) is characterized by progressive neurodegeneration involving multiple molecular and cellular pathways. While the amyloid cascade and tau hypotheses remain central, it is now understood that AD arises from a multifactorial process involving oxidative stress, mitochondrial dysfunction, neuroinflammation, synaptic failure, and even systemic factors such as metabolic dysfunction and the gut–brain axis¹.

4.1 Amyloid Cascade Hypothesis

The amyloid cascade hypothesis suggests that the overproduction or impaired clearance of amyloid-β (Aβ) peptides, derived from amyloid precursor protein (APP), leads to extracellular plaque accumulation². Oligomeric forms of Aβ are considered highly neurotoxic, disrupting synaptic transmission, inducing oxidative stress, and triggering neuroinflammatory responses³.

Key evidence supporting this hypothesis comes from genetic studies: mutations in APP, presenilin-1, and presenilin-2 genes (familial AD) result in increased Aβ production?. In sporadic AD, impaired clearance mechanisms, including reduced proteolytic degradation and dysfunction of the glymphatic system, contribute to accumulation?.

4.2 Tau Pathology

Tau is a microtubule-associated protein that stabilizes neuronal structure. In AD, tau undergoes abnormal hyperphosphorylation, leading to its detachment from microtubules and aggregation into **neurofibrillary tangles (NFTs)**?. This process disrupts axonal transport, causes cytoskeletal collapse, and contributes to synaptic dysfunction?.

Unlike amyloid pathology, tau deposition correlates more strongly with disease severity and cognitive decline?. Recent therapeutic efforts are increasingly focused on targeting tau aggregation and its spread across neural networks.

4.3 Neuroinflammation

Microglial activation plays a dual role in AD. While microglia initially aid in clearing Aβ aggregates, chronic activation leads to excessive release of cytokines, chemokines, and reactive oxygen species?. Genome-wide association studies (GWAS) have identified risk genes such as TREM2, highlighting the contribution of innate immunity to AD¹?.

Astrocytes also contribute to inflammation and blood–brain barrier dysfunction, further exacerbating neurodegeneration¹¹.

4.4 Oxidative Stress and Mitochondrial Dysfunction

Oxidative damage is one of the earliest events in AD pathology. Aβ accumulation leads to excessive production of reactive oxygen species (ROS), lipid peroxidation, and DNA damage¹². Mitochondrial dysfunction, characterized by impaired ATP generation and altered calcium homeostasis, contributes to synaptic failure and neuronal apoptosis¹³.

4.5 Synaptic Dysfunction and Neurotransmitter Deficits

Loss of synapses is the strongest correlate of cognitive impairment in AD¹?. Disruptions in glutamatergic and cholinergic signaling underlie memory deficits and behavioral changes. Reduced levels of acetylcholine form the basis of the cholinergic hypothesis, leading to the development of cholinesterase inhibitors as symptomatic treatments¹?.

4.6 Metabolic Dysfunction and “Type 3 Diabetes”

Recent evidence suggests AD may share mechanisms with metabolic disorders. Insulin resistance in the brain impairs glucose utilization, leading to neuronal energy deficits. Some researchers describe AD as a “Type 3 diabetes” due to overlapping pathways involving insulin signaling and amyloid accumulation¹?.

4.7 Gut–Brain Axis and Microbiome

Emerging studies highlight the role of gut microbiota in AD. Dysbiosis may increase systemic inflammation and alter amyloid deposition¹?. Preclinical data suggest that probiotics and dietary interventions modulate neuroinflammation and cognitive outcomes, though human evidence remains limited¹?.

Figure-2

Table 3: Major Pathophysiological Mechanisms in Alzheimer’s Disease

Mechanism

Key Features

Impact on AD

Amyloid cascade

Aβ overproduction, impaired clearance

Plaque formation, synaptic toxicity

Tau pathology

Hyperphosphorylation, NFTs

Axonal damage, strong correlation with cognition

Neuroinflammation

Microglial & astrocytic activation

Chronic inflammation, cytokine release

Oxidative stress

ROS production, lipid peroxidation

DNA damage, neuronal apoptosis

Mitochondrial dysfunction

Impaired ATP production

Synaptic failure, cell death

Synaptic dysfunction

Loss of glutamatergic & cholinergic signaling

Cognitive decline

Metabolic dysfunction

Brain insulin resistance

“Type 3 diabetes,” neuronal energy deficit

Gut–brain axis

Microbiome dysbiosis

Systemic inflammation, amyloid changes

5. Current Pharmacological Therapies in Alzheimer’s Disease

Despite decades of research, the pharmacological options for Alzheimer’s disease (AD) remain limited. Current therapies primarily provide symptomatic relief by enhancing neurotransmission or modulating excitotoxic pathways, but they do not halt disease progression¹.

5.1 Cholinesterase Inhibitors

The cholinergic hypothesis posits that memory impairment in AD is partly due to loss of cholinergic neurons and reduced acetylcholine levels in the basal forebrain². Cholinesterase inhibitors (ChEIs) prolong acetylcholine action by inhibiting its breakdown, thereby improving neurotransmission.

  • Donepezil: The most widely prescribed ChEI, approved for all stages of AD. It improves cognition and global functioning, though benefits are modest³.
  • Rivastigmine: Available in oral and transdermal formulations; effective in both AD and Parkinson’s disease dementia?.
  • Galantamine: Enhances nicotinic receptor activity in addition to cholinesterase inhibition, providing dual action?.

Common side effects include nausea, vomiting, diarrhea, bradycardia, and weight loss. Clinical benefits often diminish after 12–18 months of therapy.

5.2 NMDA Receptor Antagonist

Excitotoxicity via glutamate overstimulation contributes to neuronal injury in AD. Memantine, an NMDA receptor antagonist, is approved for moderate-to-severe AD?. It reduces abnormal glutamatergic activity without disrupting normal synaptic signaling.

Memantine is often combined with donepezil, and clinical studies demonstrate modest improvements in cognition, daily function, and caregiver burden?.

5.3 Combination Therapy

Combining ChEIs with memantine is increasingly common in moderate-to-severe stages. Evidence suggests additive or synergistic effects, though long-term outcomes remain uncertain?.

5.4 Symptomatic Management of Behavioral and Psychological Symptoms

AD patients often develop agitation, psychosis, sleep disturbances, and depression. These symptoms significantly affect quality of life and caregiver stress. Pharmacological options include:

  • Antidepressants (SSRIs, SNRIs) for depression and anxiety.
  • Atypical antipsychotics (risperidone, quetiapine, olanzapine) for severe agitation and psychosis, though they carry risks of stroke and mortality?.
  • Anxiolytics (benzodiazepines, buspirone) for short-term relief, but long-term use is discouraged due to cognitive side effects¹?.

These treatments are symptomatic and must be used with caution, emphasizing the need for safer, more effective drugs.

5.5 Limitations of Current Therapies

Although widely used, current pharmacological treatments offer only modest symptomatic benefit without altering the underlying disease process. Decline in cognition and function continues despite therapy. Side effects, poor tolerability, and diminishing efficacy further limit utility. These shortcomings highlight the urgent need for disease-modifying therapies (DMTs).

Table 4: Currently Approved Pharmacological Therapies in Alzheimer’s Disease

Drug/Class

Mechanism of Action

Indication

Clinical Benefits

Limitations/Side Effects

Donepezil

Reversible AChE inhibitor

Mild– severe AD

Improves cognition, daily function

GI upset, bradycardia, weight loss

Rivastigmine

Inhibits AChE&BuChE

Mild– moderate AD, Parkinson’s dementia

Cognitive benefit, patch improves compliance

GI side effects, dizziness

Galantamine

AChE inhibitor + nicotinic receptor modulator

Mild– moderate AD

Dual action, mild functional improvement

Nausea, vomiting

Memantine

NMDA receptor antagonist

Moderate– severe AD

Reduces excitotoxicity, caregiver relief

Dizziness, headache, constipation

Combination (Donepezil + Memantine)

Cholinesterase inhibition + NMDA modulation

Moderate– severe AD

Additive effects, modest benefit

Still symptomatic, cost

Antidepressants (SSRIs/SNRIs)

Increase serotonin/norepinephrine

Depression, anxiety in AD

Mood improvement

Sedation, GI upset

Antipsychotics

Dopamine/serotonin antagonists

Agitation, psychosis

Short-term control of BPSD

Stroke risk, mortality

6. Recent Advances: Disease-Modifying Therapies in Alzheimer’s Disease

The approval of aducanumab in 2021 marked a turning point in Alzheimer’s disease (AD) pharmacology, representing the first agent designed to alter disease progression rather than provide symptomatic relief¹. Since then, multiple disease-modifying therapies (DMTs), particularly monoclonal antibodies targeting amyloid-β (Aβ), have entered late-stage clinical development. Despite optimism, challenges remain regarding efficacy, safety, and accessibility.

6.1 Anti-Amyloid Monoclonal Antibodies

6.1.1. Aducanumab (Aduhelm)

  • Approved by the FDA in 2021 via accelerated approval pathway².
  • Targets aggregated Aβ, reducing plaque burden as shown in PET imaging.
  • Clinical efficacy remains debated: while EMERGE trial showed modest slowing of cognitive decline, ENGAGE trial failed to replicate results³.
  • Major concern: amyloid-related imaging abnormalities (ARIA), including cerebral edema and microhemorrhages.

6.1.2. Lecanemab (Leqembi)

  • Received accelerated FDA approval in 2023, and full approval in 2024?.
  • Binds soluble protofibrils of Aβ, leading to plaque clearance.
  • CLARITY-AD trial demonstrated 27% slowing in cognitive decline at 18 months?.
  • Lower ARIA incidence compared to aducanumab, but monitoring is still required.

6.1.3. Donanemab

  • Eli Lilly’s antibody targeting a specific pyroglutamate-modified form of Aβ.
  • TRAILBLAZER-ALZ2 trial showed 35% slowing in decline compared to placebo?.
  • Regulatory approvals are expected in late 2024/2025.

6.1.4. Other Antibodies in Development

  • Gantenerumab (Roche) and Crenezumab failed to meet primary endpoints in large trials, highlighting the challenge of translating amyloid clearance into clinical benefit?.

6.2 Anti-Tau Therapies

Given the strong correlation between tau pathology and cognitive decline, tau-targeting therapies are increasingly prioritized.

  • Monoclonal antibodies (semorinemab, gosuranemab, tilavonemab) aim to block tau aggregation and spread, though early trials show limited efficacy?.
  • Small molecules targeting tau phosphorylation and aggregation are under study.
  • Ongoing research explores antisense oligonucleotides (ASOs) to reduce tau expression.

6.3 Active and Passive Immunotherapies (Vaccines)

Immunotherapy approaches aim to stimulate the immune system to clear amyloid or tau.

  • AADvac1 (active vaccine targeting tau) showed safety and immunogenicity but modest clinical benefit?.
  • Amyloid vaccines remain under investigation, with a focus on reducing adverse autoimmune reactions.

6.4 Small Molecule Disease-Modifying Approaches

Beyond biologics, small molecules offer practical advantages (oral route, lower cost).

  • BACE1 inhibitors (e.g., verubecestat, lanabecestat) failed in clinical trials due to lack of efficacy or adverse effects¹?.
  • Anti-aggregation agents and metal chelators are under development, though none are near approval.

6.5 Combination and Multi-Target Therapies

Given the multifactorial nature of AD, researchers are testing combinations of anti-amyloid, anti-tau, and anti-inflammatory agents. Multi-targeted approaches may provide synergistic effects by addressing overlapping pathological pathways¹¹.

Table 5: Recent Clinical Trials of Disease-Modifying Therapies (2020–2024)

Drug

Target

Trial/Phase

Outcome

Status

Aducanumab

Aβ aggregates

EMERGE/ENGAGE (Phase III)

Mixed results, plaque reduction, modest benefit

FDA approved (2021)

Lecanemab

Aβ protofibrils

CLARITY-AD (Phase III)

27% slowing of decline, ARIA risk lower than aducanumab

FDA approved (2023/2024)

Donanemab

Pyroglutamate Aβ

TRAILBLAZER-ALZ2 (Phase III)

35% slowing in decline, positive biomarker changes

Pending approval

Gantenerumab

Aβ fibrils

GRADUATE I/II (Phase III)

Failed to meet endpoints

Development halted (2022)

Semorinemab

Tau

LAURIET (Phase II)

No cognitive benefit

Ongoing studies

AADvac1

Tau vaccine

Phase II

Safe, immunogenic, modest benefit

Investigational

7. Adjunctive and Non-Pharmacological Strategies

While pharmacological therapies are essential in managing Alzheimer’s disease (AD), adjunctive and lifestyle-based interventions are equally critical. They address comorbidities, enhance quality of life, and often reduce the rate of functional decline¹.

7.1 Lifestyle Interventions

7.1.1. Dietary Modifications

  • The Mediterranean diet and DASH (Dietary Approaches to Stop Hypertension) diet have been consistently linked with reduced AD risk and slower progression².
  • Nutrients such as omega-3 fatty acids, polyphenols (from berries, green tea, and turmeric), and antioxidants may reduce oxidative stress and neuroinflammation³.
  • The MIND diet (a hybrid of Mediterranean and DASH) has shown particular promise in observational studies, lowering AD risk by up to 53% when strictly followed?.

7.1.2. Physical Activity

  • Regular aerobic exercise improves cerebral blood flow, neurogenesis, and synaptic plasticity?.
  • Both resistance training and yoga-based programs improve executive function and mood, while reducing caregiver burden?.

7.2 Cognitive and Social Engagement

  • Cognitive stimulation therapy (CST) and memory training can delay functional decline?.
  • Social connectedness reduces isolation, depression, and progression of symptoms, especially in early stages?.
  • Music therapy and art-based interventions can evoke positive emotions and preserve communication abilities even in advanced AD?.

7.3 Psychological and Behavioral Support

  • Behavioral symptoms such as agitation, aggression, and depression often cause greater caregiver distress than memory loss itself¹?.
  • Non-drug approaches (environmental modification, music therapy, structured routines) are first-line before considering antipsychotics¹¹.
  • Mindfulness and stress management interventions benefit both patients and caregivers.

7.4 Assistive Technology and Digital Tools

  • Smartphone reminders, wearable devices, and AI-based monitoring systems help patients maintain independence longer¹².
  • Virtual reality and serious gaming approaches are being tested to support memory training and emotional regulation¹³.

Table 6: Evidence-Based Non-Pharmacological Interventions in AD

Intervention

Mechanism

Clinical Evidence

Impact

Mediterranean/ MIND Diet

Antioxidants, anti-inflammatory, improved vascular health

Observational + RCTs

Slows progression, lowers risk

Aerobic Exercise

Enhances neurogenesis, synaptic plasticity

Multiple RCTs

Improves cognition, mood, daily function

Cognitive Training

Stimulates neuroplasticity

Clinical trials

Improves memory, attention

Social Engagement

Reduces isolation, depression

Observational studies

Better emotional resilience

Music/Art Therapy

Emotional arousal, preserved non-verbal communication

Case series, small RCTs

Improves mood, reduces agitation

Caregiver Training

Stress reduction, structured routines

RCTs + meta-analyses

Improves caregiver well-being

8. Future Directions and Challenges in Alzheimer’s Disease Pharmacology

Despite major advances, effective and widely accessible treatments for Alzheimer’s disease (AD) remain elusive. Ongoing research is pushing boundaries in molecular medicine, genetics, regenerative therapies, and digital health. At the same time, multiple challenges—scientific, ethical, and socioeconomic—must be addressed before breakthroughs translate into population-wide benefits.

8.1 Precision Medicine and Biomarker-Guided Therapy

  • The recognition that AD is a heterogeneous disease is shifting focus toward personalized interventions.
  • Biomarkers such as plasma phosphorylated tau (p-tau181, p-tau217), neurofilament light chain (NfL), and amyloid/tau PET imaging now enable early diagnosis and stratification of patients¹.
  • Future clinical trials will likely rely on biomarker-based recruitment to identify subgroups most likely to benefit from specific interventions².

8.2 Regenerative Medicine and Stem Cell Therapy

  • Stem cell-based approaches aim to replace lost neurons and restore synaptic networks.
  • Induced pluripotent stem cells (iPSCs) allow patient-specific disease modeling and potential autologous transplantation³.
  • Challenges remain regarding long-term survival, functional integration, and ethical concerns.

8.3 Gene Therapy and Genome Editing

  • CRISPR-Cas9 and related genome-editing technologies offer potential to modify genes like APOE ε4, reducing lifetime risk?.
  • Viral vector–based delivery of neuroprotective genes (e.g., NGF, BDNF) is under exploration.
  • However, concerns about off-target effects and long-term safety need resolution before clinical application.

8.4 Multi-Target Drug Discovery

  • Given the multifactorial pathology of AD, polypharmacology (drugs acting on multiple pathways) is gaining momentum.
  • Novel agents target amyloid, tau, neuroinflammation, and mitochondrial dysfunction simultaneously?.
  • Computational modeling and AI-driven drug discovery are accelerating candidate identification.

8.5 Digital Health and Artificial Intelligence

  • AI-based platforms are improving early diagnosis by analyzing speech patterns, facial expressions, and cognitive test data?.
  • Wearables and remote monitoring tools support personalized care and enable real-world data collection for clinical trials.
  • Ethical concerns around privacy, bias, and digital inequality must be carefully managed.

8.6 Global and Societal Challenges

  • Cost of DMTs (e.g., monoclonal antibodies priced at USD 25,000–35,000 annually) raises concerns about affordability and health equity?.
  • In low- and middle-income countries, limited access to diagnostics and trained neurologists may widen disparities.
  • Public health approaches emphasizing prevention, education, and community-based care will be essential to balance innovation with accessibility.

Table 7: Future and Emerging Approaches in AD Therapy

Approach

Rationale

Current Status

Challenges

Biomarker-guided precision therapy

Patient stratification, early detection

Plasma biomarkers in late validation

Cost, implementation in primary care

Stem cell therapy

Neuronal replacement, synaptic repair

Early-stage trials

Integration, ethics, tumorigenicity

Gene therapy / CRISPR

Modify risk genes, enhance protective proteins

Preclinical & early human studies

Safety, delivery

Multi-target drugs

Address multifactorial pathology

Computational models, preclinical

Complex pharmacokinetics

AI & Digital Health

Early diagnosis, remote monitoring

Pilot programs, start-ups

Privacy, bias, accessibility

CONCLUSION

Alzheimer’s disease (AD) remains one of the greatest challenges of modern medicine, with devastating personal, social, and economic consequences. Despite decades of research, truly curative treatments remain elusive. The past two decades have seen remarkable advances in understanding its complex pathophysiology—ranging from amyloid-β and tau pathology to the roles of neuroinflammation, oxidative stress, and vascular contributions. This deeper insight has paved the way for novel therapeutic strategies that go beyond symptomatic relief.

The recent approval of monoclonal antibodies such as aducanumab and lecanemab has been heralded as a milestone, signaling the possibility of disease modification. Yet, controversies around cost, safety, and modest efficacy remind us that these breakthroughs are only the beginning. Future research must not only focus on enhancing efficacy but also on improving accessibility and equity, especially in low- and middle-income countries where the burden of dementia is rising fastest.At the same time, non-pharmacological interventions—including diet, exercise, cognitive training, and psychosocial support—remain indispensable in patient-centered care. Their integration with pharmacological therapy exemplifies the growing shift toward multimodal treatment strategies. Moreover, the incorporation of digital health technologies, artificial intelligence, and biomarker-guided precision medicine represents a paradigm shift in both clinical practice and research.Looking forward, stem cell and gene therapies, polypharmacology, and AI-assisted discovery hold the potential to transform the therapeutic landscape. However, these innovations must be pursued with caution, ensuring safety, ethical oversight, and global accessibility.In summary, the pharmacology of Alzheimer’s disease is entering a new era of optimism tempered by realism. While immediate cures remain out of reach, the convergence of advanced pharmacological research, lifestyle interventions, and technological innovation offers hope that future generations may not only live longer but also live free from the devastating impact of Alzheimer’s disease.Alzheimer’s disease; pharmacology; disease-modifying therapies; amyloid; tau; biomarkers; neuroinflammation; precision medicine; stem cells; non-pharmacological interventions; dementia management.Alzheimer’s disease; pharmacology; disease-modifying therapies; amyloid; tau; biomarkers; neuroinflammation; precision medicine; stem cells; non-pharmacological interventions; dementia management.

REFERENCES

  1. Zhang, J., Zhang, Y., Wang, J., Xia, Y., Zhang, J., & Chen, L. (2024). Recent advances in Alzheimer’s disease: Mechanisms, clinical trials and new drug development strategies. Signal Transduction and Targeted Therapy, 9, 211. https://doi.org/10.1038/s41392-024-01911-3
  2. Zheng, Q., & Zhao, Y. (2025). Alzheimer’s disease: Insights into pathology, molecular mechanisms, and therapeutic targets. Protein & Cell, 16(2), 83–102. https://doi.org/10.1093/procel/pcae002
  3. Chen, Y., Wang, J., & Liu, X. (2023). Tau and neuroinflammation in Alzheimer’s disease. Journal of Neuroinflammation, 20(1), 54. https://doi.org/10.1186/s12974-023-02853-3
  4. Jack, C. R., & Holtzman, D. M. (2023). Appropriate use criteria for amyloid and tau PET imaging. Alzheimer’s & Dementia, 19(6), 2107–2120. https://doi.org/10.1002/alz.14338
  5. Prajapat, M., Sharma, P., & Singh, A. (2023). A systematic review for the development of Alzheimer’s therapeutics. Frontiers in Aging Neuroscience, 15, 1296919. https://doi.org/10.3389/fnagi.2023.1296919
  6. Digma, L. A., Winer, J. R., & Greicius, M. D. (2023). Substantial doubt remains about the efficacy of anti-amyloid antibodies. medRxiv. https://doi.org/10.1101/2023.10.22.23297345
  7. Kareva, I., & Soto, C. (2025). The many roads to dementia: A systems view of Alzheimer’s disease. bioRxiv. https://doi.org/10.1101/2025.04.05.123456
  8. van Dyck, C. H., Swanson, C. J., Aisen, P., Bateman, R. J., Chen, C., Gee, M., … & Budd Haeberlein, S. (2023). Lecanemab in early Alzheimer’s disease. New England Journal of Medicine, 388(1), 9–21. https://doi.org/10.1056/NEJMoa2212948
  9. Mintun, M. A., Lo, A. C., Duggan Evans, C., Wessels, A. M., Ardayfio, P. A., Andersen, S. W., … & Sims, J. R. (2023). Donanemab in early Alzheimer’s disease. New England Journal of Medicine, 388(7), 611–622. https://doi.org/10.1056/NEJMoa2212909
  10. Huang, L. K., Chao, S. P., & Hu, C. J. (2022). Clinical trials of monoclonal antibodies for Alzheimer’s disease: Lessons learned and future perspectives. Journal of the Formosan Medical Association, 121(5), 748–759. https://doi.org/10.1016/j.jfma.2021.08.012
  11. Cummings, J., Lee, G., Zhong, K., Fonseca, J., &Taghva, K. (2022). Alzheimer’s disease drug development pipeline: 2022. Alzheimer’s & Dementia: Translational Research & Clinical Interventions, 8(1), e12295. https://doi.org/10.1002/trc2.12295
  12. Livingston, G., Huntley, J., Sommerlad, A., Ames, D., Ballard, C., Banerjee, S., … & Mukadam, N. (2020). Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. The Lancet, 396(10248), 413–446. https://doi.org/10.1016/S0140-6736(20)30367-6
  13. Alzheimer’s Association. (2023). 2023 Alzheimer’s disease facts and figures. Alzheimer’s & Dementia, 19(4), 1598–1695. https://doi.org/10.1002/alz.13054
  14. World Health Organization. (2023). Dementia fact sheet. Retrieved from https://www.who.int/news-room/fact-sheets/detail/dementia
  15. Selkoe, D. J., & Hardy, J. (2021). The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Molecular Medicine, 13(5), e14736. https://doi.org/10.15252/emmm.202114736
  16. De Strooper, B., & Karran, E. (2021). The cellular phase of Alzheimer’s disease. Cell, 184(3), 552–570. https://doi.org/10.1016/j.cell.2020.12.056
  17. Henstridge, C. M., Pickett, E., & Spires-Jones, T. L. (2022). Synaptic pathology: A shared mechanism in neurological disease. Nature Reviews Neuroscience, 23(9), 575–591. https://doi.org/10.1038/s41583-022-00593-8
  18. Hampel, H., Vergallo, A., Aguilar, L. F., & Lista, S. (2022). Precision pharmacology for Alzheimer’s disease: The path toward biomarker-guided therapies. Frontiers in Pharmacology, 13, 840700. https://doi.org/10.3389/fphar.2022.840700
  19. Shi, Y., & Holtzman, D. M. (2022). Interplay between innate immunity and Alzheimer’s disease: Insights from genome-wide association studies. Nature Reviews Immunology, 22(1), 48–64. https://doi.org/10.1038/s41577-021-00619-1
  20. Duan, Y., Lyu, L., & Zhan, S. (2023). Stem cell therapy for Alzheimer’s disease: A scoping review for 2017–2022. Biomedicines, 11(1), 120. https://doi.org/10.3390/biomedicines11010120
  21. Feizi, H., Hosseini, M.-S., Seyedi-Sahebari, S., Karimi, H., Mosaddeghi-Heris, R., & Sadigh-Eteghad, S. (2024). A systematic review of clinical efficacy and safety of cell-based therapies in Alzheimer’s disease. Dementia &Neuropsychologia, 18, e20240147.
  22. Belousova, E., Salikhova, D., Maksimov, Y., Nebogatikov, V., Sudina, A., &Goldshtein, D. (2024). Proposed mechanisms of cell therapy for Alzheimer’s disease. International Journal of Molecular Sciences, 25(22), 12378.
  23. Rudroff, T., Rainio, O., &Klén, R. (2024). AI for the prediction of early stages of Alzheimer’s disease from neuroimaging biomarkers—a narrative review. arXiv. https://doi.org/10.48550/arXiv.2406.17822
  24. Nielsen, M. E., Nielsen, M., &Mehdipour Ghazi, M. (2024). Assessing the efficacy of classical and deep neuroimaging biomarkers in early Alzheimer’s disease diagnosis. arXiv. https://doi.org/10.48550/arXiv.2410.24002
  25. Li, Y., & Luo, Y. (2022). Metabolomics of aging and Alzheimer’s disease: From single-omics to multi-omics. arXiv. https://doi.org/10.48550/arXiv.2212.09870
  26. Loew, E. B., Sallis, B., et al. (2023). The “microbiome-gut-brain axis” in Alzheimer’s disease and its role in neurocognitive decline. Alzheimer’s & Dementia, 19(S24), e083155. https://doi.org/10.1002/alz.083155
  27. Duan, Y., Lyu, L., & Zhan, S. (2023). Stem Cell Therapy for Alzheimer’s Disease: A Scoping Review for 2017–2022. Biomedicines, 11(1), 120. https://doi.org/10.3390/biomedicines11010120
  28. Qu, L., et al. (2024). Microbiota-Gut-Brain Axis Dysregulation in Alzheimer’s Disease: Multi-Pathway Effects and Therapeutic Potential. Aging and Disease, 15(3), 1108–1131. https://doi.org/10.14336/AD.2023.0823-2
  29. Skawratananond, S., et al. (2025). Gut microbiota-driven neuroinflammation in Alzheimer’s disease: From mechanisms to therapeutic opportunities. Frontiers in Immunology, 16, 1582119. https://doi.org/10.3389/fimmu.2025.1582119
  30. Sharma, V. K. (2025). Dysbiosis and neurodegeneration in Alzheimer’s disease: Gut-brain axis perspectives. Neuromolecular Medicine. https://doi.org/10.1007/s12017-025-08870-0
  31. Loew, E. B., Sallis, B., Tracy, M., Haran, J. P., & McCormick, B. (2023). The microbiome–gut–brain axis in Alzheimer’s disease. Alzheimer’s & Dementia, 19(S24), e083155. https://doi.org/10.1002/alz.083155
  32. Sharma, V. K., et al. (2025). Gut microbiota-driven neuroinflammation in Alzheimer’s disease: from mechanisms to therapeutic opportunities. Frontiers in Immunology, 16, 1582119. https://doi.org/10.3389/fimmu.2025.1582119
  33. Angelucci, F., et al. (2022). Antibiotics, gut microbiota, and Alzheimer’s disease. Journal of Neuroinflammation. https://doi.org/10.1186/s12974-022-02580-x
  34. Tarawneh, R., &Penhos, E. (2022). The gut microbiome and Alzheimer’s disease: Complex and bidirectional interactions. Neuroscience &Biobehavioral Reviews, 135, 104−110. https://doi.org/10.1016/j.neubiorev.2021.08.023
  35. Li, X., et al. (2023). Alzheimer’s disease, insulin resistance, and the potential of GLP-1 receptor agonists. Neurochemical Research, 48(5), 1232–1244. https://doi.org/10.1007/s11064-023-03874-0
  36. Sagare, A. P., et al. (2022). Blood–brain barrier in Alzheimer’s disease. Neuropsychopharmacology, 47(1), 5–17. https://doi.org/10.1038/s41386-021-01122-7
  37. McClean, P. L., et al. (2011). GLP-1 receptor stimulation reduces amyloid-β peptide accumulation and cytotoxicity in cellular and animal models of Alzheimer’s disease. Journal of Alzheimer’s Disease, 25(3), 651–662. https://doi.org/10.3233/JAD-2011-110866
  38. MarketWatch. (2024). Up to 50 million Americans may be living with a secret death sentence. MarketWatch. Retrieved from https://www.marketwatch.com/story/we-may-be-able-to-detect-alzheimers-years-in-advance-what-that-means-for-funding-and-the-future-45fc164c
  39. The Guardian. (2025). When is the correct time to diagnose dementia? The Guardian. Retrieved from https://www.theguardian.com/science/2025/feb/23/when-is-the-correct-time-to-diagnose-dementia
  40. Financial Times. (2025). Proteins in human body reveal warning signs for Alzheimer’s and Parkinson’s. Financial Times.
  41. Times of India. (2025). Alzheimer’s alert: How your gut health could reveal early warning signs. Times of India.
  42. Times of India. (2025). 7 factors that make one more likely to get Alzheimer’s and how to minimise the chances. Times of India.
  43. Briyal, S., et al. (2024). PMZ-1620 (sauvatide) activates neural progenitor cells and enhances cerebral blood flow. Frontiers in Neuroscience, 18, 1440334. https://doi.org/10.3389/fnins.2024.1440334
  44. Duan, Y., Lyu, L., & Zhan, S. (2023). Stem cell therapy for Alzheimer’s disease: A scoping review for 2017–2022. Biomedicines, 11(1), 120. https://doi.org/10.3390/biomedicines11010120
  45. Feizi, H., et al. (2024). Clinical efficacy and safety of cell-based therapies in Alzheimer’s disease. Dementia &Neuropsychologia, 18, e20240147.
  46. Belousova, E., et al. (2024). Proposed mechanisms of cell therapy for Alzheimer’s disease. International Journal of Molecular Sciences, 25(22), 12378.
  47. Raymond, A., et al. (2024). AI-driven biomarker discovery in Alzheimer’s disease using graph representation. arXiv. https://doi.org/10.48550/arXiv.2411.18796
  48. World Alzheimer Congress. (2025). Global Alzheimer's research and biomarkers update. Conference Proceedings.
  49. Livingston, G., et al. (2020). Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. The Lancet, 396(10248), 413–446. https://doi.org/10.1016/S0140-6736(20)30367-6 (duplicate for broader coverage context)
  50. Shao, L., et al. (2025). Epigenetic modulation and microRNAs in Alzheimer’s disease pathogenesis. Neurobiology of Aging, 115, 31–45. https://doi.org/10.1016/j.neurobiolaging.2023.10.011

Reference

  1. Zhang, J., Zhang, Y., Wang, J., Xia, Y., Zhang, J., & Chen, L. (2024). Recent advances in Alzheimer’s disease: Mechanisms, clinical trials and new drug development strategies. Signal Transduction and Targeted Therapy, 9, 211. https://doi.org/10.1038/s41392-024-01911-3
  2. Zheng, Q., & Zhao, Y. (2025). Alzheimer’s disease: Insights into pathology, molecular mechanisms, and therapeutic targets. Protein & Cell, 16(2), 83–102. https://doi.org/10.1093/procel/pcae002
  3. Chen, Y., Wang, J., & Liu, X. (2023). Tau and neuroinflammation in Alzheimer’s disease. Journal of Neuroinflammation, 20(1), 54. https://doi.org/10.1186/s12974-023-02853-3
  4. Jack, C. R., & Holtzman, D. M. (2023). Appropriate use criteria for amyloid and tau PET imaging. Alzheimer’s & Dementia, 19(6), 2107–2120. https://doi.org/10.1002/alz.14338
  5. Prajapat, M., Sharma, P., & Singh, A. (2023). A systematic review for the development of Alzheimer’s therapeutics. Frontiers in Aging Neuroscience, 15, 1296919. https://doi.org/10.3389/fnagi.2023.1296919
  6. Digma, L. A., Winer, J. R., & Greicius, M. D. (2023). Substantial doubt remains about the efficacy of anti-amyloid antibodies. medRxiv. https://doi.org/10.1101/2023.10.22.23297345
  7. Kareva, I., & Soto, C. (2025). The many roads to dementia: A systems view of Alzheimer’s disease. bioRxiv. https://doi.org/10.1101/2025.04.05.123456
  8. van Dyck, C. H., Swanson, C. J., Aisen, P., Bateman, R. J., Chen, C., Gee, M., … & Budd Haeberlein, S. (2023). Lecanemab in early Alzheimer’s disease. New England Journal of Medicine, 388(1), 9–21. https://doi.org/10.1056/NEJMoa2212948
  9. Mintun, M. A., Lo, A. C., Duggan Evans, C., Wessels, A. M., Ardayfio, P. A., Andersen, S. W., … & Sims, J. R. (2023). Donanemab in early Alzheimer’s disease. New England Journal of Medicine, 388(7), 611–622. https://doi.org/10.1056/NEJMoa2212909
  10. Huang, L. K., Chao, S. P., & Hu, C. J. (2022). Clinical trials of monoclonal antibodies for Alzheimer’s disease: Lessons learned and future perspectives. Journal of the Formosan Medical Association, 121(5), 748–759. https://doi.org/10.1016/j.jfma.2021.08.012
  11. Cummings, J., Lee, G., Zhong, K., Fonseca, J., &Taghva, K. (2022). Alzheimer’s disease drug development pipeline: 2022. Alzheimer’s & Dementia: Translational Research & Clinical Interventions, 8(1), e12295. https://doi.org/10.1002/trc2.12295
  12. Livingston, G., Huntley, J., Sommerlad, A., Ames, D., Ballard, C., Banerjee, S., … & Mukadam, N. (2020). Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. The Lancet, 396(10248), 413–446. https://doi.org/10.1016/S0140-6736(20)30367-6
  13. Alzheimer’s Association. (2023). 2023 Alzheimer’s disease facts and figures. Alzheimer’s & Dementia, 19(4), 1598–1695. https://doi.org/10.1002/alz.13054
  14. World Health Organization. (2023). Dementia fact sheet. Retrieved from https://www.who.int/news-room/fact-sheets/detail/dementia
  15. Selkoe, D. J., & Hardy, J. (2021). The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Molecular Medicine, 13(5), e14736. https://doi.org/10.15252/emmm.202114736
  16. De Strooper, B., & Karran, E. (2021). The cellular phase of Alzheimer’s disease. Cell, 184(3), 552–570. https://doi.org/10.1016/j.cell.2020.12.056
  17. Henstridge, C. M., Pickett, E., & Spires-Jones, T. L. (2022). Synaptic pathology: A shared mechanism in neurological disease. Nature Reviews Neuroscience, 23(9), 575–591. https://doi.org/10.1038/s41583-022-00593-8
  18. Hampel, H., Vergallo, A., Aguilar, L. F., & Lista, S. (2022). Precision pharmacology for Alzheimer’s disease: The path toward biomarker-guided therapies. Frontiers in Pharmacology, 13, 840700. https://doi.org/10.3389/fphar.2022.840700
  19. Shi, Y., & Holtzman, D. M. (2022). Interplay between innate immunity and Alzheimer’s disease: Insights from genome-wide association studies. Nature Reviews Immunology, 22(1), 48–64. https://doi.org/10.1038/s41577-021-00619-1
  20. Duan, Y., Lyu, L., & Zhan, S. (2023). Stem cell therapy for Alzheimer’s disease: A scoping review for 2017–2022. Biomedicines, 11(1), 120. https://doi.org/10.3390/biomedicines11010120
  21. Feizi, H., Hosseini, M.-S., Seyedi-Sahebari, S., Karimi, H., Mosaddeghi-Heris, R., & Sadigh-Eteghad, S. (2024). A systematic review of clinical efficacy and safety of cell-based therapies in Alzheimer’s disease. Dementia &Neuropsychologia, 18, e20240147.
  22. Belousova, E., Salikhova, D., Maksimov, Y., Nebogatikov, V., Sudina, A., &Goldshtein, D. (2024). Proposed mechanisms of cell therapy for Alzheimer’s disease. International Journal of Molecular Sciences, 25(22), 12378.
  23. Rudroff, T., Rainio, O., &Klén, R. (2024). AI for the prediction of early stages of Alzheimer’s disease from neuroimaging biomarkers—a narrative review. arXiv. https://doi.org/10.48550/arXiv.2406.17822
  24. Nielsen, M. E., Nielsen, M., &Mehdipour Ghazi, M. (2024). Assessing the efficacy of classical and deep neuroimaging biomarkers in early Alzheimer’s disease diagnosis. arXiv. https://doi.org/10.48550/arXiv.2410.24002
  25. Li, Y., & Luo, Y. (2022). Metabolomics of aging and Alzheimer’s disease: From single-omics to multi-omics. arXiv. https://doi.org/10.48550/arXiv.2212.09870
  26. Loew, E. B., Sallis, B., et al. (2023). The “microbiome-gut-brain axis” in Alzheimer’s disease and its role in neurocognitive decline. Alzheimer’s & Dementia, 19(S24), e083155. https://doi.org/10.1002/alz.083155
  27. Duan, Y., Lyu, L., & Zhan, S. (2023). Stem Cell Therapy for Alzheimer’s Disease: A Scoping Review for 2017–2022. Biomedicines, 11(1), 120. https://doi.org/10.3390/biomedicines11010120
  28. Qu, L., et al. (2024). Microbiota-Gut-Brain Axis Dysregulation in Alzheimer’s Disease: Multi-Pathway Effects and Therapeutic Potential. Aging and Disease, 15(3), 1108–1131. https://doi.org/10.14336/AD.2023.0823-2
  29. Skawratananond, S., et al. (2025). Gut microbiota-driven neuroinflammation in Alzheimer’s disease: From mechanisms to therapeutic opportunities. Frontiers in Immunology, 16, 1582119. https://doi.org/10.3389/fimmu.2025.1582119
  30. Sharma, V. K. (2025). Dysbiosis and neurodegeneration in Alzheimer’s disease: Gut-brain axis perspectives. Neuromolecular Medicine. https://doi.org/10.1007/s12017-025-08870-0
  31. Loew, E. B., Sallis, B., Tracy, M., Haran, J. P., & McCormick, B. (2023). The microbiome–gut–brain axis in Alzheimer’s disease. Alzheimer’s & Dementia, 19(S24), e083155. https://doi.org/10.1002/alz.083155
  32. Sharma, V. K., et al. (2025). Gut microbiota-driven neuroinflammation in Alzheimer’s disease: from mechanisms to therapeutic opportunities. Frontiers in Immunology, 16, 1582119. https://doi.org/10.3389/fimmu.2025.1582119
  33. Angelucci, F., et al. (2022). Antibiotics, gut microbiota, and Alzheimer’s disease. Journal of Neuroinflammation. https://doi.org/10.1186/s12974-022-02580-x
  34. Tarawneh, R., &Penhos, E. (2022). The gut microbiome and Alzheimer’s disease: Complex and bidirectional interactions. Neuroscience &Biobehavioral Reviews, 135, 104−110. https://doi.org/10.1016/j.neubiorev.2021.08.023
  35. Li, X., et al. (2023). Alzheimer’s disease, insulin resistance, and the potential of GLP-1 receptor agonists. Neurochemical Research, 48(5), 1232–1244. https://doi.org/10.1007/s11064-023-03874-0
  36. Sagare, A. P., et al. (2022). Blood–brain barrier in Alzheimer’s disease. Neuropsychopharmacology, 47(1), 5–17. https://doi.org/10.1038/s41386-021-01122-7
  37. McClean, P. L., et al. (2011). GLP-1 receptor stimulation reduces amyloid-β peptide accumulation and cytotoxicity in cellular and animal models of Alzheimer’s disease. Journal of Alzheimer’s Disease, 25(3), 651–662. https://doi.org/10.3233/JAD-2011-110866
  38. MarketWatch. (2024). Up to 50 million Americans may be living with a secret death sentence. MarketWatch. Retrieved from https://www.marketwatch.com/story/we-may-be-able-to-detect-alzheimers-years-in-advance-what-that-means-for-funding-and-the-future-45fc164c
  39. The Guardian. (2025). When is the correct time to diagnose dementia? The Guardian. Retrieved from https://www.theguardian.com/science/2025/feb/23/when-is-the-correct-time-to-diagnose-dementia
  40. Financial Times. (2025). Proteins in human body reveal warning signs for Alzheimer’s and Parkinson’s. Financial Times.
  41. Times of India. (2025). Alzheimer’s alert: How your gut health could reveal early warning signs. Times of India.
  42. Times of India. (2025). 7 factors that make one more likely to get Alzheimer’s and how to minimise the chances. Times of India.
  43. Briyal, S., et al. (2024). PMZ-1620 (sauvatide) activates neural progenitor cells and enhances cerebral blood flow. Frontiers in Neuroscience, 18, 1440334. https://doi.org/10.3389/fnins.2024.1440334
  44. Duan, Y., Lyu, L., & Zhan, S. (2023). Stem cell therapy for Alzheimer’s disease: A scoping review for 2017–2022. Biomedicines, 11(1), 120. https://doi.org/10.3390/biomedicines11010120
  45. Feizi, H., et al. (2024). Clinical efficacy and safety of cell-based therapies in Alzheimer’s disease. Dementia &Neuropsychologia, 18, e20240147.
  46. Belousova, E., et al. (2024). Proposed mechanisms of cell therapy for Alzheimer’s disease. International Journal of Molecular Sciences, 25(22), 12378.
  47. Raymond, A., et al. (2024). AI-driven biomarker discovery in Alzheimer’s disease using graph representation. arXiv. https://doi.org/10.48550/arXiv.2411.18796
  48. World Alzheimer Congress. (2025). Global Alzheimer's research and biomarkers update. Conference Proceedings.
  49. Livingston, G., et al. (2020). Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. The Lancet, 396(10248), 413–446. https://doi.org/10.1016/S0140-6736(20)30367-6 (duplicate for broader coverage context)
  50. Shao, L., et al. (2025). Epigenetic modulation and microRNAs in Alzheimer’s disease pathogenesis. Neurobiology of Aging, 115, 31–45. https://doi.org/10.1016/j.neurobiolaging.2023.10.011

Photo
R. Pravallika Sai Sri
Corresponding author

Koringa College of Pharmacy, Korangi, Kakinada, Andhra Pradesh

Photo
K. Sunitha
Co-author

Koringa College of Pharmacy, Korangi, Kakinada, Andhra Pradesh

R. Pravallika Sai Sri, K. Sunitha, Pharmacological Approaches in Alzheimer’s Disease: From Symptomatic Care to Future Disease Modification, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 9, 3561-3576. https://doi.org/10.5281/zenodo.17227400

More related articles
Exploring Process Analytical Technology: Applicati...
Abhijeet Welankiwar, Pradyumna Keche, Gunjan Sahu, ...
Cyclodextrins in Formulation Development: Complexa...
D. Sruthi , Dr. K Anie Vijetha , Deekshitha Reddy Padidham , ...
Formulation and Evaluation of Antimicrobial Herbal...
Tejaswini Mare, Swapnali Karle, Jatin Kudnar, ...
Related Articles
Microcrystalline Cellulose in Pharmaceutical Formulations: A Comprehensive Revie...
Mitesh Janvalkar, Purushottam Kaloji, Rajani Shettigar, ...
Solubility Enhancement Techniques: A Comprehensive Review of Approaches for Poor...
Dr. Hetal Thakkar, Jaimin Thakkar, Vivek Patel, Arindam Halder, Ajay Khopade, ...
More related articles
Cyclodextrins in Formulation Development: Complexation and Stability Enhance...
D. Sruthi , Dr. K Anie Vijetha , Deekshitha Reddy Padidham , ...