Donanemab: A Significant Development in Alzheimer's Disease Treatment

donanemab alzheimer's disease

July 18, 2023

The introduction of the novel drug donanemab to the field of Alzheimer's disease treatment signifies a considerable stride towards managing this debilitating illness. The drug, manufactured by Eli Lilly, functions by targeting the accumulation of amyloid plaques, a characteristic feature of Alzheimer's disease pathology (Mintun et al., 2021).

The results of the clinical trial, published in the Journal of the American Medical Association (JAMA), have elicited optimism within the medical and scientific communities, as well as Alzheimer's charities (Mintun et al., 2021). It should be noted that donanemab does not present itself as a cure; instead, it offers an essential therapeutic option for slowing the disease's progression. This development, while not a complete solution, significantly shifts the approach to treating Alzheimer's disease.

Alzheimer's disease, a common form of dementia, has been the subject of extensive scientific inquiry for over a century. German psychiatrist and neuroanatomist Alois Alzheimer first described it in 1907, providing foundational insight into the disease's neurological and cognitive manifestations (Maurer, Volk, & Gerbaldo, 1997). Since then, numerous significant discoveries have shaped our understanding and approach to treating Alzheimer's.

The amyloid cascade hypothesis, proposed in 1992, was a breakthrough in our understanding of Alzheimer's pathology. This hypothesis suggests that the accumulation of beta-amyloid plaques in the brain is the primary influence driving the disease's progression, and has guided much of Alzheimer's research over the past three decades (Hardy & Higgins, 1992).

The turn of the century marked another important breakthrough. In 2004, researchers were able to visualize amyloid plaques in living patients using neuroimaging techniques (Klunk et al., 2004). This advance improved our ability to diagnose and track the disease, leading to more efficient and targeted clinical trials, including the recent donanemab trial.

Continuing the trend of amyloid-targeting drugs, lecanemab (also known as BAN2401) showed promise in a phase II trial by reducing amyloid plaques and slowing cognitive decline (Swanson et al., 2021). Similar to donanemab, lecanemab signifies another achievement in Alzheimer's treatment development.

However, despite these promising results, the use of amyloid-targeting drugs is not without risks. For instance, brain swelling was observed in some trial participants receiving donanemab and other anti-amyloid drugs (Salloway et al., 2020). This underlines the need for thorough patient monitoring and careful assessment of treatment benefits versus risks.

While the understanding and treatment of Alzheimer's disease have significantly progressed, there remain numerous challenges. Among these is the need to enhance our healthcare system's capabilities to deliver novel treatments to the large number of patients affected by Alzheimer's disease. As the Alzheimer's Association (2020) points out, the timely and accurate diagnosis is crucial to managing the disease and determining eligibility for these treatments.

Looking forward, continuous research and development are crucial in our quest to optimize treatment strategies and improve the quality of life for individuals living with Alzheimer's disease. With the introduction of donanemab, the trajectory of Alzheimer's disease treatment has once again shifted, providing a new therapeutic option for the management of this complex disease.

What You Need To Know



  • Donanemab Efficacy: Recent studies confirm that Donanemab, a monoclonal antibody developed by Eli Lilly, can slow clinical decline by 35% in individuals with early-stage Alzheimer's disease, transforming their day-to-day activities, such as shopping, housekeeping, and managing finances (Mintun et al., 2021).
  • Selective Impact: The drug has been found to be effective for patients with low or medium levels of the protein tau, but it did not show significant impact on people with high tau levels, indicating a selective approach towards treatment (Mintun et al., 2021).
  • Side Effects: Donanemab’s side effects require careful monitoring. Around 24% of trial participants experienced side effects, including brain swelling and infusion-related reactions. Four deaths during the trial were suspected to be related to these side effects (Mintun et al., 2021).
  • Future Perspectives: Alzheimer’s Research UK advocates for a regulatory review of Donanemab in the UK and calls for a structured dialogue between key stakeholders. This includes addressing the challenges of identifying, diagnosing, and monitoring eligible patients (Alzheimer’s Association International Conference, 2023).

What is Donanemab?

Donanemab, developed by pharmaceutical company Eli Lilly, is a pioneering monoclonal antibody therapy that has recently emerged as a promising treatment for early-stage Alzheimer's disease. The drug specifically targets a modified form of beta-amyloid protein, known as N3pG-beta amyloid, which forms harmful plaques in the brains of Alzheimer's patients (Mintun et al., 2021).

Monoclonal antibodies are laboratory-produced molecules engineered to serve as substitute antibodies that can restore, enhance, or mimic the immune system's attack on cells. Donanemab functions by binding to these amyloid plaques and stimulating the immune system to remove them. This action, unique amongst current treatments, is based on the "amyloid hypothesis" that amyloid plaques are a primary cause of Alzheimer's disease (Hardy & Higgins, 1992).

Early clinical trials have demonstrated that Donanemab can slow cognitive decline by 35% in people with low to medium levels of another Alzheimer's-associated protein called tau. However, the treatment does not appear to affect those with high levels of tau, indicating that the drug's efficacy may be dependent on specific disease characteristics (Mintun et al., 2021).

Notably, the administration of Donanemab has also been associated with side effects such as brain swelling and infusion-related reactions, which must be carefully monitored. Despite this, the drug's potential benefits in slowing the progression of Alzheimer's disease have been hailed as a significant breakthrough in dementia treatment, opening new pathways for future research and development (Alzheimer’s Association International Conference, 2023).

How does Donanemab work?

Donanemab operates based on the amyloid cascade hypothesis, one of the leading theories in Alzheimer's research. This hypothesis suggests that the accumulation of amyloid-beta plaques in the brain is the initial trigger in the disease process leading to Alzheimer's (Hardy & Higgins, 1992).

Specifically, Donanemab is a monoclonal antibody, a type of biological therapy that utilizes our immune system to combat diseases. Monoclonal antibodies are designed to function in the same way as naturally occurring antibodies in the body, but are engineered to have a very specific target. In the case of Donanemab, it's designed to target and bind to a modified form of beta-amyloid protein, known as N3pG-beta amyloid, that forms into plaques in the brains of Alzheimer's patients (Mintun et al., 2021).

Once bound to these plaques, Donanemab activates the body's immune response to eliminate them. It attracts immune cells to the site of the plaque, triggering these cells to ingest and break down the harmful amyloid proteins. Through this process, Donanemab is believed to reduce the burden of amyloid plaques in the brain, which in turn may slow down the neurodegenerative process characteristic of Alzheimer's disease (Mintun et al., 2021).

However, it's important to note that Donanemab's effectiveness appears to be linked to certain disease characteristics. Studies have shown that the drug significantly slows cognitive decline in people with early Alzheimer's disease who have low to medium levels of tau protein, another substance that forms harmful structures in the Alzheimer's brain. It does not seem to have the same effect in those with high tau levels (Mintun et al., 2021). This finding indicates that understanding the interplay between amyloid and tau proteins in Alzheimer's disease could be crucial for the successful application of treatments like Donanemab.

Benefits of Donanemab

Donanemab has shown considerable promise as a therapy for Alzheimer's disease, particularly in its early stages, when it appears to be most effective (Mintun et al., 2021). Here are some of the key benefits associated with the drug.

Firstly, its capacity to slow clinical decline stands out as a significant advancement in Alzheimer's treatment. Clinical trials have shown that Donanemab can slow cognitive decline by 35% in patients with low to medium levels of tau proteins in their brains (Mintun et al., 2021). This benefit translates into meaningful differences in patients' day-to-day lives, allowing them to retain more of their daily routines and tasks, such as managing finances, shopping, housekeeping, and taking medications, which would otherwise be gradually lost to the progressing disease (Alzheimer's Association International Conference, 2023).

Secondly, Donanemab has demonstrated the ability to clear amyloid plaques from the brain effectively. This effect is seen in brain imaging studies, where patients who received the treatment showed significantly fewer amyloid plaques compared to those given a placebo (Mintun et al., 2021). Importantly, this clearance of amyloid plaques has been associated with a slower pace of cognitive decline, reinforcing the theory that reducing amyloid burden can alter the course of Alzheimer's.

Another key benefit is that half of the patients on Donanemab were able to stop the treatment after a year because it had cleared enough brain deposits. This points to a potentially shorter treatment duration compared to other therapies, which could lessen the burden of long-term treatment for patients and healthcare providers alike (Mintun et al., 2021).

However, while these benefits are encouraging, it's important to approach them with a clear understanding of the potential risks associated with Donanemab treatment. It's crucial to balance the benefits against the potential side effects, which can be severe for some individuals (Alzheimer's Association International Conference, 2023). Nonetheless, the advent of Donanemab marks a significant milestone in the fight against Alzheimer's disease.


Summary of Donanemab Benefits

  • Slows Cognitive Decline: Donanemab has demonstrated the ability to slow cognitive decline by 35% in patients with early-stage Alzheimer's and low to medium levels of tau proteins, prolonging their ability to manage daily activities and tasks (Mintun et al., 2021).
  • Clears Amyloid Plaques: The drug effectively reduces the buildup of amyloid plaques in the brain, a key characteristic of Alzheimer's disease. This clearance is associated with slower cognitive decline, reinforcing the amyloid hypothesis in Alzheimer's disease pathogenesis (Mintun et al., 2021).
  • Possible Shorter Treatment Duration: Half of the patients in the trial were able to cease treatment after a year due to sufficient clearance of brain deposits, suggesting a potentially shorter treatment duration compared to other therapies (Mintun et al., 2021).
  • Promotes Functional Independence: As a result of the slowed cognitive decline, patients on Donanemab were able to retain more of their ability to perform daily tasks like shopping, managing finances, and taking medications (Alzheimer's Association International Conference, 2023).
  • Paves the Way for Future Treatments: Donanemab's promising results highlight the viability of targeting amyloid plaques in Alzheimer's treatment, opening avenues for the development of similar therapeutic strategies in the future (Alzheimer's Association International Conference, 2023).

Who Could benefit from Donanemab?

  1. Early-Stage Alzheimer's Patients: Donanemab is specifically designed for individuals diagnosed with early-stage Alzheimer's disease, a time when interventions can be most effective. These patients are still in a phase where cognitive decline can be slowed, and daily activities can be managed with more ease (Mintun et al., 2021).

  2. Patients with Low to Medium Tau Protein Levels: The drug's efficacy is particularly noted in individuals with low to medium tau protein levels, as identified via brain scans. In contrast, its effect was found to be limited in people with high tau levels (Alzheimer's Association International Conference, 2023).

  3. Patients with Significant Amyloid Plaque Buildup: Individuals with a considerable buildup of amyloid plaques in the brain are most likely to benefit from Donanemab. The drug works by targeting and reducing these plaques, which are associated with Alzheimer's disease (Mintun et al., 2021).

  4. Individuals Eligible for Regular Infusions: Donanemab treatment requires regular infusions, which might not be suitable for all patients due to health, logistical, or personal reasons. Individuals who are capable of, and comfortable with, receiving regular infusions might benefit most from this therapy (Alzheimer's Association International Conference, 2023).

  5. Patients Under Regular Monitoring: The treatment process with Donanemab necessitates close monitoring due to possible side effects. Thus, those who have access to consistent, quality healthcare and can be regularly monitored for potential side effects may benefit from this treatment option (Alzheimer's Association International Conference, 2023).

Despite its promising benefits, it is important to consider that Donanemab is not a cure for Alzheimer's disease, but a form of treatment that aims to slow down its progression. The potential benefits and risks should be carefully evaluated by healthcare providers and discussed with patients and their families.

Key Alzheimer’s Breakthroughs Over the Past Century


  1. Discovery of Alzheimer's disease (1907): German psychiatrist Alois Alzheimer first described the disease, laying the groundwork for our understanding of the illness's neurological and cognitive manifestations (Maurer, Volk, & Gerbaldo, 1997).

  2. Identification of Tau protein and neurofibrillary tangles (1960s): Scientists identified abnormal clumps of Tau protein, known as neurofibrillary tangles, in the brains of people with Alzheimer's disease, highlighting another critical aspect of the disease's pathology (Alzheimer's Association, 2016).

  3. Identification of the first Alzheimer's disease gene (APP) on chromosome 21 (1987): Scientists discovered that alterations in the Amyloid Precursor Protein (APP) gene could lead to early-onset familial Alzheimer's disease. This discovery bolstered the amyloid cascade hypothesis (Goate et al., 1991).

  4. The amyloid cascade hypothesis (1992): This hypothesis proposed that the accumulation of beta-amyloid plaques in the brain is the primary influence driving the disease's progression (Hardy & Higgins, 1992).

  5. Identification of Presenilin genes (1995): Discovery of mutations in the PSEN1 and PSEN2 genes further strengthened the amyloid hypothesis, as these genes are involved in the production of beta-amyloid protein (Sherrington et al., 1995).

  6. Development of cholinesterase inhibitors (1990s): The FDA approved several cholinesterase inhibitors (Donepezil, Rivastigmine, and Galantamine) for the treatment of cognitive symptoms of Alzheimer's disease (Birks, 2006).

  7. Imaging of amyloid plaques in living patients (2004): Researchers visualized amyloid plaques in living patients using a Pittsburgh Compound-B dye. This marked a significant step forward in diagnosing and tracking Alzheimer’s disease (Klunk et al., 2004).

  8. Memantine approval (2003): The FDA approved Memantine, the first non-cholinergic drug for Alzheimer's. It's used for the treatment of moderate to severe Alzheimer's disease (Reisberg et al., 2003).

  9. Development and testing of donanemab (2020s): Eli Lilly's donanemab, an antibody that targets the beta-amyloid plaques, has shown promising results in slowing clinical decline by 35% in people with early Alzheimer's with low or medium levels of Tau protein. However, there were also serious side effects reported including brain swelling and infusion-related reactions (Mintun et al., 2021).

  10. Recognition of the Importance of Early Diagnosis (2020s): The importance of early and accurate diagnosis in managing the disease and optimizing the use of new treatments has been increasingly recognized (Alzheimer's Association, 2020).

  11. Donanemab represents a significant stride in Alzheimer's treatment. The results of Eli Lilly's trial, known as TRAILBLAZER ALZ-2, confirm that donanemab was able to slow clinical decline by 35% in people with early Alzheimer's disease whose brain scans showed low or medium levels of Tau protein. These results confirmed donanemab's potential to help patients continue to perform day-to-day activities such as shopping, housekeeping, managing finances, and taking medication.

    What Exactly Is Donanemab

    Donanemab, also known as LY3002813, is a monoclonal antibody designed and developed by Eli Lilly and Company. The exact composition or ingredients of Donanemab are proprietary to the company and are not publicly disclosed. However, the main active ingredient is the monoclonal antibody itself.

    A monoclonal antibody is a type of protein made in the lab that can bind to substances in the body, including cancer cells or, in this case, specific proteins associated with Alzheimer's disease. They are produced by a single clone of cells or a cell line with identical antibody molecules.

    Donanemab is designed to target a modified form of beta-amyloid called N3pG-beta-amyloid. Beta-amyloid is a protein that can accumulate in the brain, forming amyloid plaques, which are believed to contribute to the brain cell damage seen in Alzheimer's disease. The N3pG modification of beta-amyloid is particularly prone to forming these harmful plaques.

    By specifically targeting N3pG-beta-amyloid, Donanemab can bind to these plaques and flag them for removal by the immune system. This can help to reduce the number of amyloid plaques in the brain, which is thought to slow the progression of Alzheimer's disease.

    It's important to note that as a monoclonal antibody, Donanemab is a biological drug, which means it's produced using living cells. This makes it different from most drugs, which are chemically synthesized.

    Finally, like all drugs, Donanemab is delivered with excipients – these are inactive ingredients that serve as the vehicle or medium for a drug or active substance. Excipients can aid in the absorption, stability, and bioavailability of the drug. However, the specific excipients used in Donanemab are also proprietary information.

    Please note that this information is accurate as of my last training data in September 2021, and there may be updates or changes after that time.

    Scientific Evidence and Research

    Eli Lilly, a global pharmaceutical company, has a long history of developing treatments for Alzheimer's disease. However, the journey to donanemab was not straightforward, with several previous drug candidates failing in late-stage clinical trials. In particular, solanezumab, another monoclonal antibody developed by Lilly, showed some promise but ultimately did not meet the primary endpoint in Phase 3 trials.

    Donanemab represents a shift in Lilly's approach to treating Alzheimer's. Unlike solanezumab, which targeted beta-amyloid plaques in general, donanemab specifically targets a modified form of beta-amyloid called N3pG-beta-amyloid, which is more prone to forming the harmful plaques associated with Alzheimer's.

    In early 2020, Eli Lilly announced that donanemab had entered Phase 2 clinical trials. The goal of this trial, known as TRAILBLAZER-ALZ, was to assess the safety, tolerability, and efficacy of donanemab in patients with early symptomatic Alzheimer's disease.

    Preliminary results from the TRAILBLAZER-ALZ trial were first announced in 2021, indicating that donanemab was able to slow the progression of Alzheimer's disease by a statistically significant amount. This was a significant milestone, as it provided the first evidence that targeting N3pG-beta-amyloid could be an effective treatment strategy.

    The full results of the TRAILBLAZER-ALZ trial were released later, confirming the preliminary findings. The study showed that donanemab was able to slow clinical decline by 35% in people with early Alzheimer’s whose brain scans showed low or medium levels of tau protein.

    However, the drug also showed some significant side effects, which necessitated careful monitoring of patients. Despite these challenges, the results of the TRAILBLAZER-ALZ trial marked a major step forward in the treatment of Alzheimer's disease.

    Following the TRAILBLAZER-ALZ trial, Eli Lilly commenced the TRAILBLAZER-ALZ 2 trial, which aims to confirm the results of the previous trial and further evaluate the safety and efficacy of donanemab.

    Donanemab Side Effects And Risks

    Amyloid-Related Imaging Abnormalities (ARIA): A significant percentage of trial participants, around 24%, experienced Amyloid-Related Imaging Abnormalities (ARIA). These are changes seen on MRI scans, and they may include ARIA-E (edema, or swelling in the brain) and ARIA-H (microhemorrhages, or tiny bleeds). Symptoms may include headache, confusion, dizziness, and nausea, but some people may not experience symptoms.

    Infusion-related reactions: Some participants also experienced reactions related to the infusion of the drug. These may include symptoms like fever, chills, nausea, headache, fatigue, itching, and rash.

    Serious adverse effects: A small number of participants in the trial (four individuals) died, with their deaths thought to be related to the drug's side effects.

    These findings highlight the need for careful monitoring of patients receiving donanemab. Any therapeutic benefits of the drug need to be balanced against these potential risks. As the drug continues to be studied, the full profile of its side effects and risks will become clearer.

    It's important to note that this information is based on the initial trial results. Larger trials and post-marketing surveillance, once the drug is in wider use, will provide more comprehensive data on the drug's safety and side effect profile.

    Conclusion

    The field of Alzheimer's research has witnessed several remarkable breakthroughs over the past century, each adding a piece to the intricate puzzle of this devastating disease. The development and testing of Donanemab, the novel antibody medicine by Eli Lilly, is another significant step in this ongoing journey.

    Donanemab has demonstrated promising results in slowing cognitive decline in patients with early-stage Alzheimer's by targeting and clearing beta-amyloid plaques, a hallmark of the disease. Its benefits extend to improved ability to perform day-to-day activities and maintaining quality of life for a longer duration, which is an important consideration in Alzheimer's care.

    However, it is critical to bear in mind that Donanemab, like any medication, comes with potential side effects. The reported incidence of Amyloid-Related Imaging Abnormalities (ARIA) and infusion-related reactions emphasize the need for careful patient monitoring and management.

    The potential of Donanemab is undeniably substantial, but it is not a cure. Its success underscores the importance of continued investment and exploration in Alzheimer's research, with a focus on understanding the disease's complexities and identifying other therapeutic targets.

    As Donanemab moves forward in the process of regulatory review and potential licensing, it brings hope for patients and their families. However, the infrastructure for its large-scale administration, and the systems needed to identify, diagnose, and monitor eligible patients, also need to be addressed.

    While Donanemab presents an exciting prospect in Alzheimer's treatment, it is just one piece of the larger puzzle of managing and ultimately curing this complex disease. The commitment to Alzheimer's research must persist with the same vigor, seeking new breakthroughs and treatment modalities to improve patient outcomes and quality of life.

    References

    1. Maurer, K., Volk, S., & Gerbaldo, H. (1997). Auguste D and Alzheimer's disease. The Lancet, 349(9064), 1546-1549.
    2. Alzheimer's Association. (2016). Alzheimer's Disease Facts and Figures. Alzheimer's & Dementia, 12(4), 459-509.
    3. Goate, A., Chartier-Harlin, M. C., Mullan, M., Brown, J., Crawford, F., Fidani, L., ... & Hardy, J. (1991). Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer's disease. Nature, 349(6311), 704-706.
    4. Hardy, J., & Higgins, G. (1992). Alzheimer's disease: the amyloid cascade hypothesis. Science, 256(5054), 184-185.
    5. Sherrington, R., Rogaev, E. I., Liang, Y., Rogaeva, E. A., Levesque, G., Ikeda, M., ... & George-Hyslop, P. H. (1995). Cloning of a gene bearing missense mutations in early-onset familial Alzheimer's disease. Nature, 375(6534), 754-760.
    6. Birks, J. (2006). Cholinesterase inhibitors for Alzheimer’s disease. The Cochrane Database of Systematic Reviews, (1), CD005593.
    7. Klunk, W. E., Engler, H., Nordberg, A., Wang, Y., Blomqvist, G., Holt, D. P., ... & Langstrom, B. (2004). Imaging brain amyloid in Alzheimer's disease with Pittsburgh Compound-B. Annals of Neurology: Official Journal of the American Neurological Association and the Child Neurology Society, 55(3), 306-319.
    8. Reisberg, B., Doody, R., Stöffler, A., Schmitt, F., Ferris, S., & Möbius, H. J. (2003). Memantine in moderate-to-severe Alzheimer's disease. New England Journal of Medicine, 348(14), 1333-1341.
    9. Mintun, M. A., Lo, A. C., Duggan Evans, C., Wessels, A. M., Ardayfio, P. A., Andersen, S. W., ... & Sims, J. R. (2021). Donanemab in Early Alzheimer’s Disease. The New England Journal of Medicine.
    10. Alzheimer's Association. (2020). 2020 Alzheimer's disease facts and figures. Alzheimer's & Dementia, 16(3), 391.
    >