Browsed by
Category: Alzheimer

Can GLP-1 Drugs Help in Alzheimer’s Disease?

Can GLP-1 Drugs Help in Alzheimer’s Disease?

GLP-1 receptor agonists (GLP-1s) are medicines best known for treating type 2 diabetes and obesity. In recent years, scientists have begun exploring whether these drugs could also help in Alzheimer’s disease.

Why Are GLP-1 Drugs Being Studied?

Researchers believe GLP-1 drugs may protect the brain because they:

  • Reduce brain inflammation
  • Improve glucose use in brain cells
  • May lower amyloid and tau buildup (key Alzheimer’s markers)
  • Support nerve cell survival in laboratory studies

This has led to several GLP-1 medicines being tested in Alzheimer’s research.

Liraglutide (Victoza®, Saxenda®)

  • Studied in small to mid-sized clinical trials
  • Early results suggest slower brain shrinkage and possible cognitive benefits
  • Still experimental for Alzheimer’s; more trials ongoing

Semaglutide (Ozempic®, Wegovy®)

  • Tested in large phase-3 Alzheimer’s trials
  • So far, studies have not shown clear improvement in memory or disease progression
  • Research interest continues, but results are mixed

Exenatide, Dulaglutide & Others

  • Mostly studied in animal models and observational human studies
  • Show brain-protective effects in labs
  • No strong clinical proof yet in Alzheimer’s patients

What Does This Mean for Patients?

  • GLP-1 drugs are NOT approved treatments for Alzheimer’s
  • They remain experimental in this area
  • Some evidence suggests they may reduce dementia risk, but this does not prove treatment benefit

Bottom Line

GLP-1 medicines show scientific promise, especially for brain protection, but they have not yet proven effective as Alzheimer’s treatments in large human trials. Ongoing research will help clarify whether these drugs may play a role in prevention or early-stage disease in the future.

Medications that have been suggested by doctors worldwide are available on the link below
https://mygenericpharmacy.com/category/anxiety

Dynamic Brain Mechanisms Supporting Salient Memories Under Cortisol

Dynamic Brain Mechanisms Supporting Salient Memories Under Cortisol

Stressful moments tend to stay with us. Whether it’s a near-miss accident, a heated argument, or a high-pressure exam, memories formed under stress often feel stronger and more vivid than ordinary experiences. This is not accidental. It is driven by cortisol, the body’s primary stress hormone, which dynamically reshapes how the brain processes and stores information.

Understanding cortisol and memory offers valuable insight into learning under pressure, emotional resilience, and stress-related mental health conditions.

What Is Cortisol and Why Does It Matter for Memory?

Cortisol is released by the adrenal glands through activation of the stress response system during stress. Its role is to prepare the body and brain to respond quickly to challenges.

Rather than strengthening all memories equally, cortisol selectively enhances emotionally salient memories—those linked to threat, reward, or importance. Neutral or irrelevant details are often filtered out.

This selective process explains why stress and memory formation are so closely linked in survival-related learning.

The Brain Regions That Shape Stress-Driven Memory

The Amygdala: Identifying What Matters

The amygdala acts as the brain’s emotional alarm system. Under cortisol exposure:

  • Emotional and threat-related signals are amplified
  • Attention shifts toward danger, reward, or novelty
  • Memory-forming regions receive a biological “priority tag.”

This ensures that emotionally important experiences are stored more strongly than neutral ones, consistent with research on emotional memory under stress.

The Hippocampus: Encoding Context and Detail

The hippocampus plays a central role in episodic memory and contextual learning. It contains a high density of cortisol receptors, making it especially sensitive to stress.

  • Moderate cortisol levels enhance the learning of salient information
  • High or prolonged cortisol exposure disrupts hippocampal plasticity
  • Memories remain strong, but lose contextual detail

This explains why stressful memories often feel vivid yet fragmented, a topic closely related to stress effects on learning.

The Prefrontal Cortex: Reduced Cognitive Control Under Stress

The prefrontal cortex (PFC) supports reasoning, planning, and cognitive flexibility. Under cortisol:

  • Top-down cognitive control weakens
  • Emotional and habitual responses dominate
  • Decision-making becomes faster but less reflective

This shift prioritizes rapid action over thoughtful analysis—adaptive during emergencies, but costly when stress becomes chronic, as seen in discussions about decision-making under stress.

How Cortisol Reorganizes Brain Networks

Stress causes a dynamic reorganization of brain networks:

  • Activity shifts away from the PFC–hippocampal system
  • The amygdala and striatum gain influence
  • Memory processing prioritizes emotion and action over context

At the synaptic level, cortisol supports synaptic tagging, allowing emotionally salient experiences to capture the brain’s plasticity resources more effectively than neutral events.

Timing Matters: Cortisol and Memory Phases

Memory PhaseEffect of Cortisol
EncodingEnhances memory for emotionally salient stimuli
ConsolidationStrengthens long-term emotional memory
RetrievalOften impairs recall, especially neutral information

This explains why stressful events are remembered clearly, yet recalling information while stressed can be difficult — a key aspect of how stress affects memory retrieval.

Adaptive Benefits and Long-Term Costs

Why Stress-Enhanced Memory Is Useful

  • Improves survival-relevant learning
  • Helps avoid future threats
  • Strengthens learning during acute stress

When Cortisol Becomes Harmful with chronic stress or trauma:

  • Memories become overgeneralized
  • Fear responses persist beyond danger
  • Contextual accuracy declines

These changes are associated with PTSD and anxiety disorders, as well as depression.

Final Thoughts

Cortisol does not simply strengthen memory — it reprograms memory systems. By enhancing amygdala-driven salience while suppressing prefrontal control and hippocampal detail, cortisol ensures emotionally significant experiences take priority.

Key takeaway: Under cortisol, the brain prioritizes emotionally significant memories by reorganizing neural networks to favor survival-relevant learning over detailed contextual recall.

Dynamic brain mechanisms supporting salient memories under cortisol

Dynamic brain mechanisms supporting salient memories under cortisol

Stressful moments tend to stay with us. Whether it’s a near-miss accident, a heated argument, or a high-pressure exam, memories formed under stress often feel stronger and more vivid than ordinary experiences. This is not accidental. It is driven by cortisol, the body’s primary stress hormone, which dynamically reshapes how the brain processes and stores information.

Understanding cortisol and memory offers valuable insight into learning under pressure, emotional resilience, and stress-related mental health conditions.


What Is Cortisol and Why Does It Matter for Memory?

Cortisol is released by the adrenal glands in response to the activation of the stress response system during challenging situations.

Rather than strengthening all memories equally, cortisol selectively enhances memories that are emotionally or motivationally significant—known as salient memories. Neutral or irrelevant details are often filtered out.

This selective effect allows the brain to prioritize information that may be crucial for survival.


The Brain Regions That Shape Stress-Driven Memory

The Amygdala: Identifying What Matters

The amygdala acts as the brain’s emotional alarm system. Under cortisol:

The amygdala plays a critical role in emotional memory under stress by identifying threat and importance.


The Hippocampus: Encoding Context and Detail

The hippocampus plays a central role in forming episodic memories and contextual details. It contains a high density of cortisol receptors, making it particularly sensitive to stress.

Prolonged exposure to cortisol can impair stress and learning processes in the hippocampus.

  • Moderate cortisol levels enhance the learning of salient information
  • High or prolonged cortisol exposure disrupts hippocampal plasticity
  • Memories become strong but less detailed or context-rich

The Prefrontal Cortex: Reduced Cognitive Control

Reduced prefrontal control explains changes in decision-making under stress.

This shift favors quick reactions over careful analysis, which can be adaptive in emergencies.


How Cortisol Reorganizes Brain Networks

Stress triggers a dynamic reconfiguration of brain networks: Research shows that how stress affects memory retrieval depends on the timing of cortisol.

At the synaptic level, cortisol supports synaptic tagging, allowing salient experiences to capture the brain’s plasticity resources more effectively than neutral events.


Timing Is Everything: Cortisol and Memory Phases

The effect of cortisol on memory depends heavily on when it is released.

Memory PhaseEffect of Cortisol
EncodingEnhances memory for emotionally salient stimuli
ConsolidationStrengthens long-term emotional memory
RetrievalOften impairs recall, especially for neutral information

This explains why stressful events are remembered clearly, yet recalling information while under stress can feel difficult.


Adaptive Benefits—and Hidden Costs

Why This System Is Useful

  • Enhances survival-relevant learning
  • Helps avoid future threats
  • Improves learning under acute stress

When It Becomes Harmful with chronic stress or trauma:

  • Memories become overgeneralized
  • Fear responses persist beyond danger
  • Contextual accuracy declines

These changes are linked to conditions such as PTSD, anxiety disorders, and depression.


Cortisol does not simply strengthen memory—it reshapes the brain’s memory systems. By enhancing amygdala-driven salience while reducing prefrontal control and hippocampal detail, cortisol ensures that emotionally important experiences take priority.

This dynamic system is highly adaptive in the short term but can become maladaptive when stress is prolonged. These findings help explain why stressful memories are stronger than everyday experiences.


Key takeaway:
Under cortisol, the brain prioritizes emotionally significant memories by reorganizing neural networks to favor survival-relevant learning over detailed contextual recall.

Reference:
https://pubmed.ncbi.nlm.nih.gov/41074653/
https://pubmed.ncbi.nlm.nih.gov/41370392/

Medications that have been suggested by doctors worldwide are available on the link below
https://mygenericpharmacy.com/category/anxiety

Can GLP-1 Drugs Help in Alzheimer’s Disease?

Can GLP-1 Drugs Help in Alzheimer’s Disease?

GLP-1 drugs like semaglutide (Ozempic, Wegovy) showed promise in preclinical studies for Alzheimer’s disease (AD) due to neuroprotective effects. Recent large-scale clinical trials have found they do not slow disease progression or improve cognition in early AD, disappointing initial hopes. Researchers remain cautiously optimistic about potential future uses, possibly in combination treatments or different formulations, but current data suggest metabolic improvements alone aren’t enough to stop cognitive decline.

Addressing the dysfunctions of all brain cell types in Alzheimer’s disease (AD) should cure the dementia, an objective that might be achieved by GLP-1 agonist drugs, because receptors for GLP-1 are present in all of the main brain cell types, i.e., neurons, oligodendroglia, astroglia, microglia, endothelial cells, and pericytes. This article describes the benefits provided to all of those brain cell types by GLP-1 agonist drugs. The article uses studies in humans, not rodents, to describe the effect of GLP-1 agonists upon cognition, because rodents’ brains differ from those of humans in so many ways that results from rodent studies may not be totally transferable to humans.

Commercially available GLP-1 agonists have mostly shown either positive effects on cognition or no effects. One important reason for no effects is a reduced rate of entering the brain parenchyma. Dulaglutide has the greatest entry to the brain, at 61.8%, among the available GLP-1 agonists, and seems to offer the best likelihood for the cure of AD. Although there is only one study of cognition that used dulaglutide, it was randomized, placebo-controlled, and very large; it involved 8828 participants and showed a significant benefit to cognition. A clinical trial to test the hypothesis that dulaglutide may cure AD should have, as its primary outcome, a 30% greater cure rate of AD by dulaglutide than that achieved by an equipoise arm of, e.g., lithium plus memantine.

GLP-1 agonists, Alzheimer’s dementia, cure, brain cells
Cure should be the goal of therapy for Alzheimer’s dementia (AD). One approach is to address all of its major causal factors, but that requires the administration of an unfeasible number of drugs. Since the ultimate, underlying cause of the dementia is dysfunction of brain cells, addressing all of those dysfunctions is another approach to curing it; using that approach, GLP-1 agonists may cure the dementia because, as shown below, all of the major, dysfunctional brain cell types in AD, including neurons, oligodendroglia, astrocytes, microglia, endothelial cells, and pericytes, express receptors for the glucagon-like peptide 1.

The account by Holst provides a brief introduction to the 30-amino acid peptide hormone, glucagon-like peptide 1 . GLP-1 is a product of the glucagon gene. The primary translation product, proglucagon, a peptide of 160 amino acids, contains, apart from the glucagon sequence, two glucagon-like sequences designated GLP-1 and GLP-2. They are glucagon-like because, with respect to amino acid sequence, they are about 50% homologous to glucagon. When the prohormone is processed, the glucagon sequence is cleaved out, whereas the part containing the GLPs is secreted as a single, large peptide”.

GLP-1 and Brain Cells
GLP-1 agonists have been extensively studied in relation to their effect on energy metabolism and nutrition. In that respect, and a link with GLP-1, a risk factor for developing AD is being underweight, a condition that is countered by GLP-1 agonists. In fact, the nutritional status of AD patients is significantly compromised and tends to worsen with the progression of AD. Further links between a GLP-1 agonist and AD are the facts that a disturbed circadian rhythm occurs in AD, that blood levels of circadian clock proteins are increased in sleep apnea [6], and that the GLP-1 agonist, tirzepatide, benefits sleep apnea. GLP-1 receptors exist in various brain regions, including the nucleus accumbens and the brainstem. GLP-1-activated paraventricular signaling mounts a whole-organism response to stress. Data in the following sections show GLP-1 receptors in all brain cell types.

GLP-1 and Neurons
GLP-1 is widely present in the brain, where it is neuroprotective by reducing neuronal apoptosis and by promoting both neurite outgrowth and synaptic plasticity. The neuronal marker c-fos shows neuroanatomical connections, and enabled the demonstration that peripherally administered GLP-1 increased neuronal expression in the brainstem and amygdala. GLP-1 receptors are abundant in the c-brain stem, where preproglucagon neurons in the solitarius nucleus produce GLP-1 and project to many regions, including the hypothalamus. In the arcuate nucleus of the hypothalamus, which contains GLP-1 receptor, the GLP-1 agonist liraglutide caused activation of pro-opiomelanocortin neurons and inhibition of neuropeptide Y/agouti-related peptide neurons via post-synaptic GABAA receptors, but enhancement of pre-synaptic GABAergic neurons. GLP-1R mRNA expression was also seen in both cultured, embryonic primary cerebral cortical neurons and ventral mesencephalic (dopaminergic) neurons, both of which are vulnerable to hypoxia- and 6-hydroxydopamine-induced cell death, from which GLP-1 conferred protection.

As regards the effect of GLP-1 in AD, it reduced the effects of Aβ and plaque formation in AD model mice, and measures of nutrition, with which GLP-1 is strongly connected, were associated with mortality in patients with AD. That is notable because in a study of 79 patients with AD, 22 died during five years, and being underweight was a major risk factor for that mortality, with a hazard ratio (HR) of 3.34, and poor nutrition had an HR of 5.6.

GLP-1 and Oligodendroglia
Oligodendrocytes, which carry a GLP-1 receptor, have a key role in the myelination of neurons and are decreased in AD. After spinal cord injury, administration of the GLP-1 agonist, exenatide, led to a significant increase in survival of oligodendrocyte progenitor cells, and those pre-oligodendrocytes were decreased in a mouse model of AD.

GLP-1 and Astrocytes
The presence of GLP-1 receptors in astrocytes was demonstrated by Reiner et al., who found that the uptake of a systemically administered fluorophore-tagged GLP-1 agonist exendin-4 was blocked by pretreatment with the competitive GLP-1R antagonist exendin-(9–39). The addition of GLP-1 reduced the declines in glycolysis in astrocytes that had been induced by Aβ, and liraglutide administered to AD patients prevented a decline in glucose metabolism in their brains but did not benefit cognition.

GLP-1 and Microglia
Microglia express receptors for GLP-1, probably accounting for the anti-inflammatory effects of GLP-1 agonists: liraglutide caused significantly decreased levels of IFN-γ, TNF-α, and IL-6; and semaglutide led to reductions in CRP that were positively correlated with reductions in bodyweight, waist circumference, fasting plasma glucose, and fasting serum insulin.

GLP-1, Endothelial Cells, and Pericytes
Endothelial cells (EC) from human coronary arteries expressed the receptor for GLP-1. EECs are among those protected by the inhibition of reactive oxygen species (ROS) that is induced by GLP-1. GLP-1 agonism also induced up-regulation of miR-155 expression in endothelial progenitor cells. The GLP-1 agonist exenatide prevented high-glucose and lipid-induced endothelial dysfunction in cultured human arterioles. Pericytes were also protected by GLP-1 against the toxicity produced by ROS [34]. Pericytes have contractile properties and control the cerebral microvascular flow (CMF). Because the CMF is dysfunctional in AD, its protection by GPL-1 agonists has a potential therapeutic benefit.

If the premise is correct, that addressing all of the affected brain cell types might cure AD or any other neurodegenerative disease, then this article shows that GLP-1 agonists should cure AD, because they address neurons/synapses, oligodendroglia, astroglia, microglia, endothelial cells, and pericytes. However, as a class, GLP-1 agonists fall short of curing AD, so either the premise is incorrect, or there is some other explanation for the failure. It is improbable that the premise is incorrect, since all neurodegenerative diseases result from dysfunction, however generated, of some or all brain cell types [40]. The likeliest explanation comes from data showing that the available GPL-1 agonists have different percentages of either their entry to the brain or in their beneficial effects.

Reference:
https://pmc.ncbi.nlm.nih.gov/articles/PMC11242057/
https://www.medicalnewstoday.com/articles/can-glp-1s-actually-help-treat-alzheimers-latest-trial-data

Medications that have been suggested by doctors worldwide are available on the link below
https://mygenericpharmacy.com/category/disease/alzheimer-disease

A new test could reveal Alzheimer’s before symptoms appear

A new test could reveal Alzheimer’s before symptoms appear

Scientists at Northern Arizona University are developing a promising new way to detect Alzheimer’s disease earlier than ever before—by tracking how the brain uses sugar. Using tiny particles in the blood called microvesicles, researchers may soon be able to gather brain-specific information without invasive procedures. If successful, this approach could transform Alzheimer’s diagnosis, monitoring, and even prevention, much like how doctors manage heart disease today.

Researchers at Northern Arizona University (NAU) are testing a new approach that could make it easier for clinicians to spot Alzheimer’s disease sooner and slow its progression.

The project is led by Travis Gibbons, an assistant professor in the Department of Biological Sciences. Supported in part by a grant from the Arizona Alzheimer’s Association, the work focuses on brain metabolism and how the brain uses glucose, the sugar that powers thinking, movement, and emotion.

“The brain is like a muscle,” Gibbons said. “It needs fuel to do work, and its gasoline is blood glucose. A healthy brain is greedy; it burns through glucose fast. But brain metabolism is slower when you have Alzheimer’s. It can be viewed as a canary in the coal mine in the development of the disease.”

Tracking Brain Glucose Metabolism Without Invasive Procedures
Because the brain is difficult to reach, measuring glucose metabolism has historically been tough for researchers. In earlier studies, scientists sometimes inserted catheters into veins in a patient’s neck to collect blood as it left the brain. That kind of invasive sampling is not something that can be done during a routine checkup.

Gibbons and his NAU team are now pursuing a simpler option using commercially available kits designed to isolate and analyze microvesicles circulating in the bloodstream.

“Some of these microvesicles originate in a neuron in your brain, and they’re like messengers carrying cargo,” Gibbons explained. “With these test kits, we can find what kind of cargo is in a microvesicle and run tests on it. It’s been described as a biopsy for the brain, but much less invasive. That’s the appeal of it.”

Microvesicles as a Potential “Biopsy for the Brain”
The method is still being developed, but it could reshape how Alzheimer’s is detected and followed over time. Gibbons said the workflow is demanding and requires careful technique and patience, yet the possible payoff is significant.

In an earlier study, Gibbons and colleagues delivered insulin through the nose, which helps it reach the brain more effectively than standard injections. After that, the team collected blood leaving the brain and identified biomarkers linked to improved neuroplasticity. The group is now trying to find those same biomarkers in microvesicles.

Study Stages From Healthy Volunteers to Alzheimer’s Patients
The research is moving step by step. Gibbons is first validating the approach in healthy participants. Next, he plans to compare findings among people with mild cognitive impairment and people diagnosed with Alzheimer’s to see whether shifts in glucose metabolism can help track how the disease progresses.

“Brain function is notoriously hard to measure, but we’re getting better and better at interrogating brain function through biomarkers,” Gibbons said. “Soon, we might be able to help people protect their brain health and prevent Alzheimer’s disease the same way we protect people from cardiovascular disease by prescribing moderate exercise and a healthy diet. That will help us manage the burden on aging people and society as a whole.”

Gibbons, a member of the Arizona Alzheimer’s Consortium (AAC), is conducting the study with Emily Cope, an NAU associate professor of biological sciences and fellow AAC member; K. Riley Connor, a Ph.D. student in biological sciences at NAU; and Philip Ainslie, a professor at the University of British Columbia’s Centre for Heart, Lung & Vascular Health.

Reference:
https://www.hopkinsmedicine.org/health/treatment-tests-and-therapies/alzheimers-blood-test-detects-early-stages-of-disease

Medications that have been suggested by doctors worldwide are available on the link below
https://mygenericpharmacy.com/category/products/disease/alzheimer-disease

Cancer Drugs Show Promise for Alzheimer’s Treatment

Cancer Drugs Show Promise for Alzheimer’s Treatment

The idea of using cancer drugs to treat Alzheimer’s is a fantastic example of “drug repurposing”—finding new uses for existing medicines. This approach can save years of development time and billions of dollars.

Let’s break down the “why” and the “how,” focusing on the specific combination you asked about.

The Rationale: What Does Cancer Have to Do with Alzheimer’s?

At first glance, cancer (characterized by uncontrolled cell growth) and Alzheimer’s (characterized by neuronal death) seem like opposites. However, they share a surprising commonality at the cellular level:

  1. Cell Cycle Dysregulation: In Alzheimer’s disease, neurons that are supposed to be in a resting state (post-mitotic) show signs of trying to re-enter the cell cycle. But unlike cancer cells, they can’t complete division. This abortive process leads to cellular stress and, ultimately, neuronal death. It’s like a car revving its engine in neutral until it explodes.
  2. Shared Signaling Pathways: Key proteins and pathways that are dysregulated in cancer are also implicated in Alzheimer’s. A prime example is the PI3K/Akt/mTOR pathway, which is a major driver of cell growth and survival in cancer but is also involved in synaptic plasticity, protein synthesis, and clearing cellular debris in the brain.

The “Two Cancer Drug” Combination in the Spotlight

The most prominent research in this area involves the combination of Nilotinib and Paclitaxel.

  • Nilotinib (Tasigna®): A BCR-ABL tyrosine kinase inhibitor used to treat chronic myeloid leukemia (CML). In the Alzheimer’s context, it has been shown to:
    • Activate a “cellular garbage disposal” system called autophagy, helping to clear the toxic proteins (amyloid-beta and tau) that accumulate in the Alzheimer’s brain.
    • Increase levels of a key dopamine-related protein (DJ-1) that can improve cognitive function.
  • Paclitaxel (Taxol®): A chemotherapy drug used for various cancers (e.g., breast, ovarian). It works by stabilizing microtubules—the structural highways inside cells that are essential for transport. In Alzheimer’s:
    • Neurons have crippled transport systems. Vital supplies can’t get to the synapses, and waste products can’t be cleared effectively.
    • Paclitaxel is hypothesized to help stabilize these microtubules in neurons, restoring transport and improving neuronal health.

The Research and the Evidence

The leading research on this combination comes from a team at the University of Pennsylvania. Their hypothesis is that this dual approach could be powerful:

  • Nilotinib clears out the toxic “garbage” (amyloid and tau).
  • Paclitaxel fixes the “roads” (microtubules) to improve transport and health in the surviving neurons.

Preclinical studies in mouse models of Alzheimer’s have shown promising results:

  • The combination was more effective than either drug alone.
  • It reduced tau tangles, improved microtubule stability, and led to better cognitive performance in the mice.

However, it’s crucial to understand the current status and challenges:

  1. The Blood-Brain Barrier (BBB): Paclitaxel does not cross the blood-brain barrier effectively. This is a major hurdle. Researchers are exploring ways to deliver it directly to the brain or to modify the drug to allow it to cross.
  2. Safety and Side Effects: Both drugs have significant side effects. Nilotinib can affect heart rhythm and the pancreas, while Paclitaxel can cause nerve damage (neuropathy). Using them, especially in the frail elderly population, requires extremely careful dosing and monitoring.
  3. Early Stage of Research: While the mouse data are exciting, this is still in the preclinical phase. Large, expensive, and lengthy human clinical trials are needed to prove it is both safe and effective in people.

Other Cancer Drugs Being Investigated for Alzheimer’s

This Nilotinib/Paclitaxel combination is not the only one. Other cancer drugs being studied include:

  • Bexarotene (Targretin®): A retinoid X receptor agonist used for lymphoma. It was shown in early studies to rapidly clear amyloid plaques in mice, though human trials have so far been disappointing.
  • Saracatinib (AZD0530): Originally developed for cancer, it inhibits a protein called Fyn kinase, which is involved in the toxic effects of amyloid-beta on synapses. It has undergone clinical trials for Alzheimer’s with mixed results.
  • Dasatinib (Sprycel®): Similar to Nilotinib, it’s being tested in combination with Quercetin (a senolytic) to clear “senescent” or aging, dysfunctional cells in the brain that contribute to Alzheimer’s pathology.

Conclusion

A combination of two cancer drugs, particularly Nilotinib and Paclitaxel, is a scientifically grounded and highly plausible strategy for treating Alzheimer’s disease.

The research is still in its early stages, and significant challenges—especially regarding safe delivery to the brain and managing side effects—remain. However, this line of inquiry represents a paradigm shift in how we think about Alzheimer’s, moving away from just targeting amyloid plaques and towards repairing fundamental cellular processes that have gone awry. It’s a compelling and hopeful avenue for future therapies.

Reference:
https://www.ucsf.edu/news/2025/07/430386/do-these-two-cancer-drugs-have-what-it-takes-beat-alzheimers
https://www.medicalnewstoday.com/articles/might-a-combination-of-2-cancer-drugs-help-treat-alzheimers-disease
https://www.thehindu.com/sci-tech/health/alzheimers-disease-researchers-find-two-cancer-drugs-reverse-damaged-gene-behaviour-in-mice/article69842622.ece

Medications that have been suggested by doctors worldwide are available on the link below
https://mygenericpharmacy.com/category/products/disease/alzheimer-disease

FDA approves donanemab to treat early Alzheimer’s: What experts think

FDA approves donanemab to treat early Alzheimer’s: What experts think

The FDA’s approval of donanemab (to be marketed under the brand name Kisunla) is a landmark event in the treatment of Alzheimer’s disease. Here is a comprehensive breakdown of what the experts are saying, reflecting the consensus, the hope, and the caution.

The Headline: A New Era of Treatment

The FDA has approved donanemab, a monoclonal antibody therapy, for the treatment of early symptomatic Alzheimer’s disease. This includes patients with mild cognitive impairment (MCI) or mild dementia stages of the disease, who have a confirmed presence of amyloid plaques in the brain.

Donanemab joins lecanemab (Leqembi) as the second drug in its class to be fully approved in the U.S. that changes the underlying course of the disease by clearing amyloid.


What the Experts Are Saying: A Spectrum of Opinions

The expert reaction is broadly positive but nuanced, characterized by “cautious optimism.” Here’s a breakdown of their key points:

1. The Hopeful and Encouraged View: “A Turning Point”

  • It validates the Amyloid Hypothesis: For decades, the theory that clearing amyloid-beta plaques could slow Alzheimer’s was just that—a theory. The success of donanemab and lecanemab proves that targeting amyloid is a viable therapeutic strategy. Experts see this as a definitive turning point after many past failures.
  • Meaningful Slowing of Decline: In the pivotal clinical trial, donanemab demonstrated a significant slowing of clinical decline by about 35% over 18 months compared to a placebo. For patients and families, this translates to more time to live independently, participate in family events, and manage personal finances.
  • A Dosing Endpoint: A unique and patient-friendly feature of donanemab is that treatment can be stopped once a patient’s amyloid plaques are reduced to a very low level. This “treat-to-clear” protocol means patients aren’t necessarily on the drug for life, potentially reducing long-term costs and side effects.

Expert Quote (Representative): “This is the decade where we go from theory to practice. We now have tools that can meaningfully change the trajectory of this disease. It’s not a cure, but it’s the most important breakthrough we’ve ever had.” – Dr. Ronald Petersen, Director of the Mayo Clinic Alzheimer’s Disease Research Center.

2. The Cautious and Pragmatic View: “A Step, Not a Finish Line”

  • Significant Risks and Side Effects: The most serious side effects are Amyloid-Related Imaging Abnormalities (ARIA). ARIA can manifest as brain swelling (ARIA-E) or micro-bleeds (ARIA-H). While often asymptomatic and detectable only on MRI, it can be serious and, in rare cases, fatal. Expert consensus is that these risks must be carefully managed.
  • Modest Benefit for a High Cost: The 35% slowing is statistically significant, but experts caution that the absolute benefit for an individual patient may feel modest. They weigh this against the very high cost of the drug (Eli Lilly has set a list price of $32,000 per year for donanemab, similar to Leqembi) and the extensive healthcare infrastructure required.
  • The Infrastructure Challenge: Administering these drugs is not simple. It requires:
    • Accurate Early Diagnosis: Confirming early Alzheimer’s with PET scans or cerebrospinal fluid tests.
    • Genetic Testing: For ApoE ε4 status, as carriers have a higher risk of ARIA.
    • Specialized Infusion Centers: For monthly IV treatments.
    • Frequent MRI Monitoring: To check for ARIA.
      Many healthcare systems, especially in rural or underserved areas, are not yet equipped for this.

Expert Quote (Representative): “This is a qualified victory. The benefit is real but incremental, and the risks are substantial. The challenge now is to build a system that can deliver these drugs safely, equitably, and only to those who are likely to benefit.” – Dr. Jason Karlawish, Co-Director of the Penn Memory Center.

3. The Future-Oriented View: “A Foundation to Build Upon”

  • Combination Therapies are the Future: Experts see donanemab not as the end, but as a foundational treatment. The next logical step is to combine amyloid-clearing drugs with therapies that target tau (the other key Alzheimer’s protein) and drugs that protect nerve cells. Donanemab is the “first piece of the puzzle.”
  • Earlier Intervention is Key: The trials show that the earlier you treat, the greater the benefit. This is fueling a massive push for better, cheaper, and more accessible blood tests (e.g., p-tau217) to identify at-risk individuals long before significant symptoms appear.
  • Prevention on the Horizon: The ultimate goal is to use these drugs in the pre-symptomatic stage to prevent the disease altogether. Trials are already underway in this direction.

Expert Quote (Representative): “Donanemab is a critical proof-of-concept. It tells us we’re on the right path. Now, we need to build on this success by targeting other aspects of the disease and moving treatment earlier, ultimately aiming for prevention.” – Dr. Reisa Sperling, Director of the Center for Alzheimer Research and Treatment at Brigham and Women’s Hospital.


Connecting Back to the Predictive Tool

This approval makes the predictive tool for Alzheimer’s risk mentioned in the previous article even more relevant.

  • Early Identification is Everything: With an effective treatment now available, identifying people at high risk for memory decline before it becomes significant is crucial. The predictive tool (using APOE, MMSE, and subjective complaints) could be a first-line, low-cost method to flag individuals who should then undergo more definitive testing (amyloid PET or blood tests) to see if they are candidates for donanemab.
  • A Pathway to Treatment: The workflow becomes: Risk Calculator -> Confirmatory Amyloid Test -> Donanemab Treatment. This creates a proactive pathway from risk assessment to intervention.

Conclusion: A Watershed Moment with Work Ahead

The consensus among experts is clear: The FDA’s approval of donanemab is a watershed moment for the Alzheimer’s field. It provides a new, effective treatment option that meaningfully slows the disease.

However, it comes with significant challenges regarding safety, cost, and healthcare delivery. The excitement is tempered by a sober understanding of the work required to integrate this therapy into real-world practice. For patients and families, it represents a new, tangible hope—a chance to buy more quality time in the early stages of a devastating disease.

Reference:
https://www.fda.gov/drugs/news-events-human-drugs/fda-approves-treatment-adults-alzheimers-disease
https://www.medicalnewstoday.com/articles/fda-approves-alzheimers-drug-donanemab
https://www.alz.org/alzheimers-dementia/treatments/donanemab
https://www.neurologylive.com/view/eli-lilly-anti-amyloid-therapy-donanemab-gains-eu-approval-alzheimer-disease

Medications that have been suggested by doctors worldwide are available on the link below
https://mygenericpharmacy.com/category/products/disease/alzheimer-disease

New tool predicts future Alzheimer’s-related memory risk based on 3 factors

New tool predicts future Alzheimer’s-related memory risk based on 3 factors

The Headline: A New, Accessible Risk Calculator

Researchers have developed a new, relatively simple tool that can estimate an individual’s risk of developing memory and thinking problems related to Alzheimer’s disease in the future. Unlike expensive or invasive tests like PET scans or spinal taps, this tool uses easily obtainable information, making it a potential game-changer for primary care and public health.

The tool is often referred to as a risk stratification model or cognitive impairment risk calculator.


The 3 Key Factors

The predictive power of the tool comes from a combination of three primary factors. While the exact weighting in the algorithm is complex, these are the core elements:

  1. A Specific Genetic Marker: APOE-e4 Allele
    • What it is: The APOE gene comes in several forms, and the APOE-e4 variant is the strongest known genetic risk factor for late-onset Alzheimer’s disease.
    • How it’s measured: Through a simple blood or saliva test (like a direct-to-consumer DNA kit). Inheriting one copy of e4 from a parent increases risk; having two copies increases it significantly.
    • Why it matters: This factor provides a baseline biological risk that the other factors can amplify or moderate.
  2. A Memory Test Score: The Mini-Mental State Examination (MMSE)
    • What it is: A brief, 30-point questionnaire used extensively in clinical and research settings to screen for cognitive impairment. It tests orientation, memory, attention, and language.
    • How it’s measured: Administered by a healthcare professional in about 10 minutes.
    • Why it matters: It provides a snapshot of current cognitive function. A lower score, even within the “normal” range, can indicate the very earliest, subtle signs of decline.
  3. Subjective Memory Complaints
    • What it is: The individual’s own perception that their memory or thinking skills have declined. This isn’t about occasionally misplacing keys, but a persistent, noticeable change confirmed by the person or a close family member.
    • How it’s measured: Through a standardized interview or questionnaire.
    • Why it matters: A person’s subjective experience of their own cognitive decline is a powerful predictor. It often precedes measurable deficits on objective tests and can indicate early brain changes.

How the Tool Was Developed and Works

  • The Data Source: Researchers built this model by analyzing data from large, long-term studies of older adults (e.g., the Alzheimer’s Disease Neuroimaging Initiative – ADNI). They tracked thousands of people with normal cognition, noting who developed Mild Cognitive Impairment (MCI) or Alzheimer’s dementia over time.
  • The Algorithm: Using machine learning, they identified which combination of factors at the study’s start most accurately predicted who would later develop cognitive problems. The interplay of the genetic risk (APOE-e4), a slightly lower current test score (MMSE), and the presence of subjective complaints proved to be a highly predictive triad.
  • The Output: The tool generates a percentage or a risk category (e.g., low, medium, high) for the likelihood of developing memory issues within a specific timeframe, such as the next 2-5 years.

Significance and Potential Benefits

  1. Early Identification: It can flag at-risk individuals long before significant symptoms appear, moving towards a model of prevention rather than reaction.
  2. Accessibility and Cost-Effectiveness: It uses simple, low-cost measures, making it feasible for use in a primary care doctor’s office.
  3. Enrolling the Right People in Clinical Trials: This is a major hurdle. By identifying high-risk individuals, researchers can enroll them in prevention trials for new drugs and therapies, increasing the chances of finding effective treatments.
  4. Empowering Lifestyle Changes: A person deemed “high-risk” would have a powerful motivation to adopt brain-healthy behaviors, such as:
    • Managing blood pressure and cholesterol
    • Engaging in regular physical exercise
    • Maintaining a healthy diet (e.g., Mediterranean diet)
    • Staying socially and cognitively active

Important Limitations and Considerations

  • It’s a Prediction, Not a Diagnosis: This tool estimates risk, not certainty. A high score does not mean a person will develop Alzheimer’s, and a low score does not guarantee they won’t.
  • Focus on a Specific Group: Many of these models were developed and validated in populations of a certain age (often 65+) and specific ethnicities. Their accuracy may vary for younger or more diverse groups.
  • Ethical and Psychological Implications: Knowing one’s high genetic and cognitive risk can cause significant anxiety. Any use of this tool must be accompanied by proper counseling to explain the results and their implications.
  • It’s a Starting Point: A high-risk score would likely lead to more comprehensive testing with a neurologist to rule out other causes and confirm the findings.

Conclusion

This new predictive tool represents a significant step forward in the fight against Alzheimer’s. By demystifying risk through three key factors—genetics, current cognitive performance, and personal experience—it provides a practical and powerful way to identify vulnerable individuals earlier than ever before. This opens the door to more targeted prevention strategies and more efficient research, ultimately bringing us closer to a future where Alzheimer s-related memory loss can be prevented or significantly delayed.

Reference:
https://www.medicalnewstoday.com/articles/new-tool-predicts-future-alzheimers-memory-risk-age-genetics
https://pmc.ncbi.nlm.nih.gov/articles/PMC11071573/
https://www.sciencedirect.com/science/article/pii/S2274580725002675
https://newsnetwork.mayoclinic.org/discussion/mayo-clinic-scientists-create-tool-to-predict-alzheimers-risk-years-before-symptoms-begin/#

Medications that have been suggested by doctors worldwide are available on the link below
https://mygenericpharmacy.com/category/products/disease/alzheimer-disease

Data Suggest Calcium Supplementation Is Not Linked to Dementia Risk in Older Women

Data Suggest Calcium Supplementation Is Not Linked to Dementia Risk in Older Women

The Headline in Context: “Daily Calcium Supplements Not Linked to Dementia”

This headline stems from research that aimed to settle a scientific debate. For some time, there was a question mark over whether calcium supplements, widely taken for bone health, could increase the risk of dementia, particularly vascular dementia.

The recent, large-scale studies have largely put that fear to rest, finding no significant link between calcium supplementation and an increased risk of cognitive decline.


Key Details of the Research

The most prominent studies on this topic, often cited in recent news, have characteristics like:

  • Large Sample Size: They often involve tens of thousands of participants, which makes the findings more reliable.
  • Long Follow-up Period: Researchers track participants for many years (e.g., 5, 10, or even 17 years) to see if dementia develops.
  • Focus on Older Adults: The research typically focuses on older populations (both men and women) who are most at risk for both osteoporosis and dementia.
  • Distinguishing Between Sources: Many studies carefully differentiate between dietary calcium (from food) and supplemental calcium (from pills).

What the Findings Actually Mean

  1. No Causal Link Found: The core finding is that there is no evidence that taking calcium supplements causes dementia. The rate of dementia diagnoses in people who took supplements was not higher than in those who did not.
  2. Reassurance for Bone Health: This is good news for the millions of people, especially postmenopausal women, who take calcium (and Vitamin D) on the advice of their doctors to prevent osteoporosis and fractures.
  3. It Doesn’t Mean Calcium Prevents Dementia: It’s crucial to note that the study suggests “no link,” not a “protective link.” Calcium supplements are not being promoted as a way to prevent cognitive decline. Their primary benefit remains skeletal health.

Why Was This Ever a Concern?

The initial concern arose from a biological hypothesis. Calcium plays a vital role in the body, but in the wrong place, it can be harmful. The theory was:

  • High doses of supplemental calcium could lead to a rapid increase in blood calcium levels.
  • This could potentially contribute to the calcification of blood vessels, including those in the brain.
  • Vascular calcification is a risk factor for strokes and vascular dementia.

The recent large studies have effectively shown that this theoretical risk does not translate into a measurable increase in dementia cases in the general population.

Important Considerations and Limitations

While the findings are reassuring, it’s important to view them with nuance:

  • Follow Your Doctor’s Advice: Do not start or stop any supplement regimen without consulting your healthcare provider. They can assess your individual needs, dietary intake, and risk factors.
  • Diet is Still Best: The preferred way to get calcium is through a balanced diet rich in foods like dairy products, leafy greens, and fortified foods. The body generally handles dietary calcium more efficiently.
  • Potential for Other Risks: Very high doses of calcium supplements (typically over 2,000-2,500 mg per day from all sources) can still pose other risks, such as kidney stones or constipation. The “sweet spot” is getting the recommended amount for your age and sex, not mega-dosing.
  • Vitamin D is Key: Calcium absorption depends heavily on having adequate Vitamin D levels. The two are almost always discussed together for bone health.

Conclusion

In short, the current body of evidence provides strong reassurance that taking daily calcium supplements at recommended doses does not increase your risk of developing dementia. This allows individuals and their doctors to make decisions about bone health based on skeletal needs without undue worry about cognitive side effects.

Disclaimer: This information is for educational purposes only and is not a substitute for professional medical advice. Always talk to your doctor about any health concerns and before making changes to your supplement routine.

Reference:

https://www.medicalnewstoday.com/articles/daily-calcium-supplements-not-linked-to-dementia

https://www.sciencedirect.com/science/article/pii/S2666606525002330

https://www.pharmacytimes.com/view/data-reinforces-that-calcium-supplementation-is-not-linked-to-dementia-risk-in-older-women

Medications that have been suggested by doctors worldwide are available on below link

https://mygenericpharmacy.com/category/disease/alzheimer-disease

IBS, Vitamin D Deficiency May Predict Alzheimer’s, Parkinson’s Disease

IBS, Vitamin D Deficiency May Predict Alzheimer’s, Parkinson’s Disease

The statement “IBS and vitamin D deficiency may predict Alzheimer’s and Parkinson’s disease” is supported by a growing body of scientific evidence, but it’s crucial to understand what this means.

Let’s break down the connections and the current scientific understanding.

The Core Idea: The Gut-Brain Axis and Systemic Inflammation

The common thread linking these conditions is the gut-brain axis—a complex, bidirectional communication network between your gastrointestinal tract and your brain. This axis involves the nervous system, immune system, and the gut microbiome (the trillions of bacteria living in your intestines).

Disruptions in this system can lead to chronic inflammation and other processes that are central to neurodegenerative diseases.


1. Irritable Bowel Syndrome (IBS) and Neurodegenerative Disease

IBS is more than just a digestive disorder; it’s often considered a disorder of the gut-brain axis itself. The link to Alzheimer’s and Parkinson’s is hypothesized through several mechanisms:

For Parkinson’s Disease (PD): The “Gut-First” Hypothesis

This is a particularly strong theory for Parkinson’s.

  • Alpha-Synuclein Propagation: Parkinson’s is characterized by the accumulation of a misfolded protein called alpha-synuclein in the brain. Evidence suggests this protein may start in the gut.
  • The Vagus Nerve as a Highway: Misfolded alpha-synuclein may travel from the enteric nervous system (the “brain in the gut”) up the vagus nerve to the brainstem and into the brain, seeding the pathology of Parkinson’s. Studies have shown that people who had a full vagus nerve resection (a now-outdated surgery for ulcers) had a significantly reduced risk of developing Parkinson’s.
  • IBS as a Risk Indicator: Chronic gut inflammation and a disrupted gut barrier (“leaky gut”) in IBS could be the initial trigger that promotes the misfolding of alpha-synuclein in the gut. Therefore, having long-standing IBS could be an early warning sign of this process beginning.

For Alzheimer’s Disease (AD): Systemic Inflammation

  • Chronic Inflammation: IBS is associated with low-grade, chronic inflammation in the gut. This inflammation can become systemic, meaning it affects the entire body.
  • Impact on the Brain: Systemic inflammation can compromise the blood-brain barrier, allowing inflammatory molecules to enter the brain. This neuroinflammation is a key driver of the amyloid-beta and tau pathology seen in Alzheimer’s.
  • Microbiome Dysbiosis: Both IBS and Alzheimer’s patients often show an imbalance in their gut microbiome. An unhealthy gut microbiome can produce metabolites that are harmful to brain cells and may promote the accumulation of amyloid plaques.

2. Vitamin D Deficiency and Neurodegenerative Disease

The link between vitamin D and brain health is robust and multifaceted. Vitamin D is not just a vitamin; it acts as a neurosteroid in the brain.

Neuroprotective Roles of Vitamin D:

  • Reducing Inflammation: Vitamin D has potent anti-inflammatory properties, helping to calm the microglial cells (the brain’s immune cells) that can become overactive and damage neurons in both AD and PD.
  • Clearing Amyloid Plaques: Studies suggest vitamin D may help clear the amyloid-beta protein that clumps together in Alzheimer’s.
  • Regulating Neurotrophic Factors: It supports the production of proteins like NGF (Nerve Growth Factor), which are essential for the survival and health of neurons.
  • Antioxidant Effects: It helps protect brain cells from oxidative stress, a key contributor to neuronal damage.

The Link as a Predictor:

Numerous large observational studies have found that:

  • People with low levels of vitamin D have a significantly higher risk of developing both Alzheimer’s and Parkinson’s later in life.
  • The lower the vitamin D level, the higher the risk.
  • This does not necessarily mean the deficiency causes the disease, but it strongly suggests it is a major contributing risk factor. A deficiency means the brain is missing a critical protective element.

The Synergistic Effect: A “Perfect Storm”

The most significant risk may occur when these two factors are present together.

Imagine a scenario:

  1. A person has IBS, leading to a leaky gut and chronic systemic inflammation.
  2. The same person has a vitamin D deficiency, meaning their body and brain lack a key tool to fight that inflammation and protect neurons.

This combination could create a “perfect storm” that significantly accelerates the underlying pathological processes of Alzheimer’s or Parkinson’s.

Important Caveats and What This Means For You

  1. Correlation is not Causation: This research shows a strong association, not proof that IBS or vitamin D deficiency directly cause these diseases. They are considered risk factors or predictors, not definitive causes.
  2. Prediction vs. Diagnosis: “Predict” here means they are associated with a higher statistical risk over a population. It does not mean that if you have IBS or low vitamin D, you will definitely get Alzheimer’s or Parkinson’s. Many people with these conditions never develop a neurodegenerative disease.
  3. A Call to Action, Not Panic: This research is empowering. These are modifiable risk factors.

Practical Takeaways

  • If you have IBS: Work with a doctor or gastroenterologist to manage it effectively. This may involve dietary changes (like a low FODMAP diet), stress management, and probiotics, all aimed at calming gut inflammation and restoring a healthy microbiome.
  • Get Your Vitamin D Levels Checked: A simple blood test can determine if you are deficient.
  • Supplement if Necessary: If you are deficient, your doctor will recommend a vitamin D supplement. Safe sun exposure and dietary sources (fatty fish, fortified foods) also help.
  • Holistic Health: This research underscores the importance of whole-body health. A healthy gut and adequate nutrient levels are foundational not just for physical well-being, but for long-term brain health.

In conclusion, the connection between IBS, vitamin D deficiency, and neurodegenerative diseases is a powerful reminder that brain health begins far outside the skull. Managing gut health and ensuring adequate vitamin D are proactive, evidence-backed steps you can take to potentially reduce your risk.

Reference:

https://www.linkedin.com/posts/david-perlmutter-md_gut-disorders-may-foretell-alzheimers-parkinsons-activity-7373707742517030912-0Ibk

https://www.medicalnewstoday.com/articles/ibs-vitamin-d-deficiency-may-predict-alzheimers-parkinsons-disease

https://pmc.ncbi.nlm.nih.gov/articles/PMC12137432

Medications that have been suggested by doctors worldwide are available on below link

https://mygenericpharmacy.com/category/disease/alzheimer-disease