1
|
Martorana A, Bonomi CG, Di Donna MG, Motta C. Lecanemab's Path Forward: Navigating the Future of Alzheimer's Treatment in Europe Amidst the EMA's Rejection. Neurol Ther 2024:10.1007/s40120-024-00675-w. [PMID: 39487946 DOI: 10.1007/s40120-024-00675-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/15/2024] [Indexed: 11/04/2024] Open
Abstract
Lecanemab (Leqembi©, Biogen), a humanized anti-amyloid-beta monoclonal antibody, has been approved for early-stage Alzheimer's disease (AD) in several countries, including the US and Japan. However, the European Medicines Agency (EMA) recently issued a negative opinion on its marketing authorization, reflecting concerns over the clinical value and manageability of anti-amyloid treatments. This decision highlights the ongoing disconnect between research advancements and clinical practice, where the focus on biological markers over tangible clinical improvements remains contentious. Despite promising biological effects, lecanemab's clinical outcomes have been modest, raising questions about its therapeutic role. The EMA's refusal underscores the need to address doubts surrounding the real-world effectiveness and safety of such treatments, especially concerning amyloid-related imaging abnormalities (ARIAs), a common side effect observed in clinical trials. The recent approval of lecanemab by the UK's Medicines and Healthcare products Regulatory Agency, despite the National Institute for Health and Care Excellence's rejection on cost-effectiveness grounds, further fuels the debate on the feasibility of anti-amyloid therapies. This commentary emphasizes the importance of real-world data on lecanemab's impact on cognitive decline, daily functioning, and side-effect management. As the global clinical use of lecanemab increases, continuous and standardized reporting on its outcomes is crucial for guiding future regulatory decisions and for potentially bridging the gap between research and practice in AD treatment.
Collapse
Affiliation(s)
- Alessandro Martorana
- Memory Clinic and Neurodegenerative Dementia Research Unit, University Hospital Policlinico Tor Vergata, University of Rome "Tor Vergata", Viale Oxford, 81, 00133, Rome, Italy.
| | - Chiara Giuseppina Bonomi
- Memory Clinic and Neurodegenerative Dementia Research Unit, University Hospital Policlinico Tor Vergata, University of Rome "Tor Vergata", Viale Oxford, 81, 00133, Rome, Italy
| | - Martina Gaia Di Donna
- Memory Clinic and Neurodegenerative Dementia Research Unit, University Hospital Policlinico Tor Vergata, University of Rome "Tor Vergata", Viale Oxford, 81, 00133, Rome, Italy
| | - Caterina Motta
- Memory Clinic and Neurodegenerative Dementia Research Unit, University Hospital Policlinico Tor Vergata, University of Rome "Tor Vergata", Viale Oxford, 81, 00133, Rome, Italy
| |
Collapse
|
2
|
Cui W, Wang Y, Tang X, Liu S, Duan Y, Gu T, Mao J, Li W, Bao J, Wei Z. CRM 197-scaffolded vaccines designed by epitope grafting ameliorate cognitive decline in an Alzheimer's disease model. Int J Biol Macromol 2024; 281:136477. [PMID: 39401639 DOI: 10.1016/j.ijbiomac.2024.136477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/20/2024]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly. Amyloid-beta (Aβ) plaque accumulation and tau neurofibrillary tangles (NFTs) formation in the brain are major neuropathological hallmarks of AD. Immunotherapies targeting Aβ and/or tau are deemed the most promising approaches for AD. Administrations with monoclonal antibodies against Aβ have yielded substantial breakthroughs clinically. Most vaccines tested clinically so far failed to prove efficacious in large part due to inappropriate design of vaccine antigens. In this study, a structure-guided approach was employed to design novel antigens targeting Aβ and/or tau by grafting multiple copies of Aβ and/or tau B-cell epitope peptide onto CRM197, which is the most widely used carrier protein in polysaccharide conjugate vaccines. The immunogenicity of the vaccines was evaluated in BALB/c mice and the efficacy was tested in a transgenic mouse model of human amyloidopathy. The antigens were highly immunogenic early vaccination substantially ameliorated cognitive decline in APP/PS1 mice and significantly reduced insoluble Aβ42/40 in the brains. These results demonstrate that the engineered antigens have protective effects on AD mice, offering a promising translatable strategy for the prevention and/or treatment of AD.
Collapse
Affiliation(s)
- Weiwei Cui
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao University, Qingdao 266071, China
| | - Ying Wang
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xiaowen Tang
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Sha Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao University, Qingdao 266071, China
| | - Yurong Duan
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao University, Qingdao 266071, China
| | - Tiantian Gu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao University, Qingdao 266071, China
| | - Junyuan Mao
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao University, Qingdao 266071, China
| | - Wenjie Li
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao University, Qingdao 266071, China
| | - Jinli Bao
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao University, Qingdao 266071, China
| | - Zhun Wei
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao University, Qingdao 266071, China; Keynova Biotech Co., Ltd., Weifang 261071, China.
| |
Collapse
|
3
|
Granzotto A, Vissel B, Sensi SL. Lost in translation: Inconvenient truths on the utility of mouse models in Alzheimer's disease research. eLife 2024; 13:e90633. [PMID: 39329365 PMCID: PMC11434637 DOI: 10.7554/elife.90633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
The recent, controversial approval of antibody-based treatments for Alzheimer's disease (AD) is fueling a heated debate on the molecular determinants of this condition. The discussion should also incorporate a critical revision of the limitations of preclinical mouse models in advancing our understanding of AD. We critically discuss the limitations of animal models, stressing the need for careful consideration of how experiments are designed and results interpreted. We identify the shortcomings of AD models to recapitulate the complexity of the human disease. We dissect these issues at the quantitative, qualitative, temporal, and context-dependent levels. We argue that these models are based on the oversimplistic assumptions proposed by the amyloid cascade hypothesis (ACH) of AD and fail to account for the multifactorial nature of the condition. By shedding light on the constraints of current experimental tools, this review aims to foster the development and implementation of more clinically relevant tools. While we do not rule out a role for preclinical models, we call for alternative approaches to be explored and, most importantly, for a re-evaluation of the ACH.
Collapse
Affiliation(s)
- Alberto Granzotto
- Center for Advanced Studies and Technology – CAST, University G. d’Annunzio of Chieti-PescaraChietiItaly
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d’Annunzio of Chieti-PescaraChietiItaly
| | - Bryce Vissel
- St Vincent’s Hospital Centre for Applied Medical Research, St Vincent’s HospitalDarlinghurstAustralia
- School of Clinical Medicine, UNSW Medicine & Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyAustralia
| | - Stefano L Sensi
- Center for Advanced Studies and Technology – CAST, University G. d’Annunzio of Chieti-PescaraChietiItaly
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d’Annunzio of Chieti-PescaraChietiItaly
- Institute for Advanced Biomedical Technologies – ITAB, University G. d’Annunzio of Chieti-PescaraChietiItaly
- Institute of Neurology, SS Annunziata University Hospital, University G. d’Annunzio of Chieti-PescaraChietiItaly
| |
Collapse
|
4
|
Daly T. A philosophy of science approach to the amyloid hypothesis of Alzheimer's disease. Eur J Neurosci 2024; 60:4707-4722. [PMID: 39119857 DOI: 10.1111/ejn.16500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/04/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
Disputes about the scientific validity of the amyloid-β hypothesis of Alzheimer's disease have been held since the early 1990s, with little constructive progress made between opposing sides despite recent therapeutic progress. Here, I argue that philosophy of science can improve the chance of constructive debate by giving researchers technical language to describe and assess scientific progress. To do so, I interpret the amyloid hypothesis using a modified version of the research programme concept from philosopher of science Imre Lakatos. I first outline the amyloid-β hypothesis and study critiques of its central place in Alzheimer's research. Then, I draw on the complexity of amyloid-β and Alzheimer's research to discuss the limits of using concepts from popular philosophers of science Karl Popper or Thomas Kuhn, before finally arguing that an adaptation of the research programme concept can foster constructive debates about the science of Alzheimer's and within it. I will argue that the amyloid-β hypothesis has contributed to significant progress in the Alzheimer's field based on what Lakatos called the "positive heuristic" (motivating the programme to test its predictions) and the "negative heuristic" (protecting the programme from refutation). I consider the amyloid research agenda to be progressive despite the fact that its claims about disease aetiology could be wrong.
Collapse
Affiliation(s)
- Timothy Daly
- Bioethics Program, FLACSO Argentina, Buenos Aires, Argentina
- Science Norms Democracy UMR 8011, Sorbonne Université, Paris, France
| |
Collapse
|
5
|
Hroudová J, Fišar Z. Alzheimer's disease approaches - Focusing on pathology, biomarkers and clinical trial candidates. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111069. [PMID: 38917881 DOI: 10.1016/j.pnpbp.2024.111069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024]
Abstract
The strategy for the development of new drugs for Alzheimer's disease (AD) recognizes that an effective therapy requires early therapeutic intervention and a multifactorial approach that considers the individual initiators of AD development. Current knowledge of AD includes the understanding of pathophysiology, risk factors, biomarkers, and the evolving patterns of biomarker abnormalities. This knowledge is essential in identifying potential molecular targets for new drug development. This review summarizes promising AD drug candidates, many of which are currently in phase 2 or 3 clinical trials. New agents are classified according to the Common Alzheimer's Disease Research Ontology (CADRO). The main targets of new drugs for AD are processes related to amyloid beta and tau neurotoxicity, neurotransmission, inflammation, metabolism and bioenergetics, synaptic plasticity, and oxidative stress. These interventions are aimed at preventing disease onset and slowing or eliminating disease progression. The efficacy of pharmacotherapy may be enhanced by combining these drugs with other treatments, antioxidants, and dietary supplements. Ongoing research into AD pathophysiology, risk factors, biomarkers, and the dynamics of biomarker abnormalities may contribute to the understanding of AD and offer hope for effective therapeutic strategies in the near future.
Collapse
Affiliation(s)
- Jana Hroudová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| | - Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| |
Collapse
|
6
|
Islam MR, Rabbi MA, Hossain T, Sultana S, Uddin S. Mechanistic Approach to Immunity and Immunotherapy of Alzheimer's Disease: A Review. ACS Chem Neurosci 2024. [PMID: 39173186 DOI: 10.1021/acschemneuro.4c00360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative condition characterized by progressive cognitive decline and memory loss, affecting millions of people worldwide. Traditional treatments, such as cholinesterase inhibitors and NMDA receptor antagonists, offer limited symptomatic relief without addressing the underlying disease mechanisms. These limitations have driven the development of more potent and effective therapies. Recent advances in immunotherapy present promising avenues for AD treatment. Immunotherapy strategies, including both active and passive approaches, harness the immune system to target and mitigate AD-related pathology. Active immunotherapy stimulates the patient's immune response to produce antibodies against AD-specific antigens, while passive immunotherapy involves administering preformed antibodies or immune cells that specifically target amyloid-β (Aβ) or tau proteins. Monoclonal antibodies, such as aducanumab and lecanemab, have shown potential in reducing Aβ plaques and slowing cognitive decline in clinical trials, despite challenges related to adverse immune responses and the need for precise targeting. This comprehensive review explores the role of the immune system in AD, evaluates the current successes and limitations of immunotherapeutic approaches, and discusses future directions for enhancing the treatment efficacy.
Collapse
Affiliation(s)
- Md Rubiath Islam
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Md Afser Rabbi
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Tanbir Hossain
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Sadia Sultana
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Shihab Uddin
- Department of Bioengineering, King Fahad University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Bio Systems and Machines, King Fahad University of Petroleum & Minerals, Dhahran-31261, Saudi Arabia
| |
Collapse
|
7
|
Underwood BR. Predicting how many people might receive treatment with new therapies for Alzheimer's disease. J Neurol Neurosurg Psychiatry 2024; 95:793. [PMID: 38862264 PMCID: PMC11347232 DOI: 10.1136/jnnp-2024-333941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/04/2024] [Indexed: 06/13/2024]
|
8
|
Gardner RS, Kyle M, Hughes K, Zhao LR. Single-Cell RNA Sequencing Reveals Immunomodulatory Effects of Stem Cell Factor and Granulocyte Colony-Stimulating Factor Treatment in the Brains of Aged APP/PS1 Mice. Biomolecules 2024; 14:827. [PMID: 39062541 PMCID: PMC11275138 DOI: 10.3390/biom14070827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) leads to progressive neurodegeneration and dementia. AD primarily affects older adults with neuropathological changes including amyloid-beta (Aβ) deposition, neuroinflammation, and neurodegeneration. We have previously demonstrated that systemic treatment with combined stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) (SCF+G-CSF) reduces the Aβ load, increases Aβ uptake by activated microglia and macrophages, reduces neuroinflammation, and restores dendrites and synapses in the brains of aged APPswe/PS1dE9 (APP/PS1) mice. However, the mechanisms underlying SCF+G-CSF-enhanced brain repair in aged APP/PS1 mice remain unclear. This study used a transcriptomic approach to identify the potential mechanisms by which SCF+G-CSF treatment modulates microglia and peripheral myeloid cells to mitigate AD pathology in the aged brain. After injections of SCF+G-CSF for 5 consecutive days, single-cell RNA sequencing was performed on CD11b+ cells isolated from the brains of 28-month-old APP/PS1 mice. The vast majority of cell clusters aligned with transcriptional profiles of microglia in various activation states. However, SCF+G-CSF treatment dramatically increased a cell population showing upregulation of marker genes related to peripheral myeloid cells. Flow cytometry data also revealed an SCF+G-CSF-induced increase of cerebral CD45high/CD11b+ active phagocytes. SCF+G-CSF treatment robustly increased the transcription of genes implicated in immune cell activation, including gene sets that regulate inflammatory processes and cell migration. The expression of S100a8 and S100a9 was robustly enhanced following SCF+G-CSF treatment in all CD11b+ cell clusters. Moreover, the topmost genes differentially expressed with SCF+G-CSF treatment were largely upregulated in S100a8/9-positive cells, suggesting a well-conserved transcriptional profile related to SCF+G-CSF treatment in resident and peripherally derived CD11b+ immune cells. This S100a8/9-associated transcriptional profile contained notable genes related to pro-inflammatory and anti-inflammatory responses, neuroprotection, and Aβ plaque inhibition or clearance. Altogether, this study reveals the immunomodulatory effects of SCF+G-CSF treatment in the aged brain with AD pathology, which will guide future studies to further uncover the therapeutic mechanisms.
Collapse
Affiliation(s)
| | | | | | - Li-Ru Zhao
- Department of Neurosurgery, State University of New York Upstate Medical University, 750 E. Adams Street, Syracuse, NY 13210, USA
| |
Collapse
|
9
|
Hierro-Bujalance C, Garcia-Alloza M. Empagliflozin reduces brain pathology in Alzheimer's disease and type 2 diabetes. Neural Regen Res 2024; 19:1189-1190. [PMID: 37905858 PMCID: PMC11467955 DOI: 10.4103/1673-5374.385865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/20/2023] [Accepted: 08/20/2023] [Indexed: 11/02/2023] Open
Affiliation(s)
- Carmen Hierro-Bujalance
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
- Salus Infirmorum-Universidad de Cadiz, Cadiz, Spain
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| |
Collapse
|
10
|
Nadeau SE. Lecanemab Questions. Neurology 2024; 102:e209320. [PMID: 38484213 DOI: 10.1212/wnl.0000000000209320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/05/2024] [Indexed: 03/19/2024] Open
Abstract
The recently published results of the 18-month randomized controlled trial of lecanemab, reporting the efficacy of the drug in slowing the progression of early Alzheimer disease, quickly led to approval by the FDA and widespread acceptance of lecanemab treatment. However, there are a number of matters that deserve further consideration. The success of blinding was not assessed, even as infusion reactions and the cerebral pathology underlying amyloid-related imaging abnormalities could have signaled to many participants that they were on drug, potentially exerting a potent placebo effect. The value of the outcome to participants is not defined in the absolute terms necessary for clinical decision-making, and the difference attributable to lecanemab was between 18% and 46% of estimates of the minimal clinically important difference on the Clinical Dementia Rating Scale Sum of Boxes. The attenuation of change on the Alzheimer's Disease Assessment Scale-Cognitive 14 achieved by lecanemab at 18 months was 50% of that achieved by donepezil at 6 months. Lecanemab treatment imposes a high treatment burden. The fact that the burden commences at the initiation of lecanemab treatment, whereas the benefit accrues years later requires us to take into account value discounting over time, which would significantly reduce the benefit/burden ratio. Finally, treatment with monoclonal antibodies to cerebral amyloid has consistently been associated with progressive cerebral atrophy. At the least, these issues should be raised in treatment discussions with patients. They also suggest a need to very seriously reconsider how we evaluate clinical trial results preparatory to translating them into clinical practice. Some suggestions are provided.
Collapse
Affiliation(s)
- Stephen E Nadeau
- From the Neurology Service and the Brain Rehabilitation Research Center, Malcom Randall VA Medical Center; Department of Neurology, University of Florida College of Medicine, Gainesville, FL
| |
Collapse
|
11
|
Afjadi MN, Dabirmanesh B, Uversky VN. Therapeutic approaches in proteinopathies. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 206:341-388. [PMID: 38811085 DOI: 10.1016/bs.pmbts.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
A family of maladies known as amyloid disorders, proteinopathy, or amyloidosis, are characterized by the accumulation of abnormal protein aggregates containing cross-β-sheet amyloid fibrils in many organs and tissues. Often, proteins that have been improperly formed or folded make up these fibrils. Nowadays, most treatments for amyloid illness focus on managing symptoms rather than curing or preventing the underlying disease process. However, recent advances in our understanding of the biology of amyloid diseases have led to the development of innovative therapies that target the emergence and accumulation of amyloid fibrils. Examples of these treatments include the use of small compounds, monoclonal antibodies, gene therapy, and others. In the end, even if the majority of therapies for amyloid diseases are symptomatic, greater research into the biology behind these disorders is identifying new targets for potential therapy and paving the way for the development of more effective treatments in the future.
Collapse
Affiliation(s)
- Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vladimir N Uversky
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Pushchino, Moscow, Russia; Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.
| |
Collapse
|
12
|
Schwinghamer K, Line S, Tesar DB, Miller DW, Sreedhara A, Siahaan TJ. Selective Uptake of Macromolecules to the Brain in Microfluidics and Animal Models Using the HAVN1 Peptide as a Blood-Brain Barrier Modulator. Mol Pharm 2024; 21:1639-1652. [PMID: 38395041 PMCID: PMC10984760 DOI: 10.1021/acs.molpharmaceut.3c00775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Monoclonal antibodies (mAbs) possess favorable pharmacokinetic properties, high binding specificity and affinity, and minimal off-target effects, making them promising therapeutic agents for central nervous system (CNS) disorders. However, their development as effective therapeutic and diagnostic agents for brain disorders is hindered by their limited ability to efficiently penetrate the blood-brain barrier (BBB). Therefore, it is crucial to develop efficient delivery methods that enhance the penetration of antibodies into the brain. Previous studies have demonstrated the potential of cadherin-derived peptides (i.e., ADTC5, HAVN1 peptides) as BBB modulators (BBBMs) to increase paracellular porosities for penetration of molecules across the BBB. Here, we test the effectiveness of the leading BBBM peptide, HAVN1 (Cyclo(1,6)SHAVSS), in enhancing the permeation of various monoclonal antibodies through the BBB using both in vitro and in vivo systems. In vitro, HAVN1 has been shown to increase the permeability of fluorescently labeled macromolecules, such as a 70 kDa dextran, 50 kDa Fab1, and 150 kDa mAb1, by 4- to 9-fold in a three-dimensional blood-brain barrier (3D-BBB) microfluidics model using a human BBB endothelial cell line (i.e., hCMEC/D3). HAVN1 was selective in modulating the BBB endothelial cell, compared to the pulmonary vascular endothelial (PVE) cell barrier. Co-administration of HAVN1 significantly improved brain depositions of mAb1, mAb2, and Fab1 in C57BL/6 mice after 15 min in the systemic circulation. Furthermore, HAVN1 still significantly enhanced brain deposition of mAb2 when it was administered 24 h after the administration of the mAb. Lastly, we observed that multiple doses of HAVN1 may have a cumulative effect on the brain deposition of mAb2 within a 24-h period. These findings offer promising insights into optimizing HAVN1 and mAb dosing regimens to control or modulate mAb brain deposition for achieving desired mAb dose in the brain to provide its therapeutic effects.
Collapse
Affiliation(s)
- Kelly Schwinghamer
- Department of Pharmaceutical Chemistry, The University of Kansas, 2093 Constant Ave., Lawrence, KS 66047, USA
| | - Stacey Line
- Department of Pharmacology and Therapeutics, University of Manitoba, 753 McDermot Avenue Winnipeg, MB, R3E 0T6, Canada
| | - Devin B. Tesar
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Donald W. Miller
- Department of Pharmacology and Therapeutics, University of Manitoba, 753 McDermot Avenue Winnipeg, MB, R3E 0T6, Canada
| | - Alavattam Sreedhara
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, The University of Kansas, 2093 Constant Ave., Lawrence, KS 66047, USA
| |
Collapse
|
13
|
Wig GS, Klausner S, Chan MY, Sullins C, Rayanki A, Seale M. Participant diversity is necessary to advance brain aging research. Trends Cogn Sci 2024; 28:92-96. [PMID: 38280836 PMCID: PMC10922549 DOI: 10.1016/j.tics.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 01/29/2024]
Abstract
An absence of population-representative participant samples has limited research in healthy brain aging. We highlight examples of what can be gained by enrolling more diverse participant cohorts, and propose recommendations for specific reforms, both in terms of how researchers accomplish this goal and how institutions support and benchmark these efforts.
Collapse
Affiliation(s)
- Gagan S Wig
- Center for Vital Longevity, The University of Texas at Dallas, Dallas, TX 75235, USA; Department of Psychology, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75080, USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Sarah Klausner
- Center for Vital Longevity, The University of Texas at Dallas, Dallas, TX 75235, USA
| | - Micaela Y Chan
- Center for Vital Longevity, The University of Texas at Dallas, Dallas, TX 75235, USA
| | - Cameron Sullins
- Center for Vital Longevity, The University of Texas at Dallas, Dallas, TX 75235, USA
| | - Anirudh Rayanki
- Department of Psychology, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75080, USA
| | - Maya Seale
- Department of Psychology, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75080, USA
| |
Collapse
|
14
|
Granzotto A, Sensi SL. Once upon a time, the Amyloid Cascade Hypothesis. Ageing Res Rev 2024; 93:102161. [PMID: 38065226 DOI: 10.1016/j.arr.2023.102161] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/23/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023]
Abstract
Recent trials with monoclonal antibodies targeting amyloid-β (Aβ) in Alzheimer's disease (AD) have sparked a renewed interest in disease-modifying therapies. Despite their promise, these trials leave the issue open and posit some doubts about the validity of the Amyloid Cascade Hypothesis (ACH). While some scores of neurocognitive tests improved upon treatment, real-world clinical benefits were minimal. This Viewpoint discusses additional, often overlooked findings from these trials. We also emphasize the multifactorial nature of AD and the need for a broader research perspective beyond the simplistic disease model provided by the ACH.
Collapse
Affiliation(s)
- Alberto Granzotto
- Center for Advanced Studies and Technology - CAST, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy; Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy.
| | - Stefano L Sensi
- Center for Advanced Studies and Technology - CAST, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy; Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy; Institute for Advanced Biomedical Technologies - ITAB, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
15
|
Roberts JA, Varma VR, Jones A, Thambisetty M. Drug Repurposing for Effective Alzheimer's Disease Medicines: Existing Methods and Novel Pharmacoepidemiological Approaches. J Alzheimers Dis 2024; 101:S299-S315. [PMID: 39422962 DOI: 10.3233/jad-240680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Drug repurposing is a methodology used to identify new clinical indications for existing drugs developed for other indications and has been successfully applied in the treatment of numerous conditions. Alzheimer's disease (AD) may be particularly well-suited to the application of drug repurposing methods given the absence of effective therapies and abundance of multi-omic data that has been generated in AD patients recently that may facilitate discovery of candidate AD drugs. A recent focus of drug repurposing has been in the application of pharmacoepidemiologic approaches to drug evaluation. Here, real-world clinical datasets with large numbers of patients are leveraged to establish observational efficacy of candidate drugs for further evaluation in disease models and clinical trials. In this review, we provide a selected overview of methods for drug repurposing, including signature matching, network analysis, molecular docking, phenotypic screening, semantic network, and pharmacoepidemiological analyses. Numerous methods have also been applied specifically to AD with the aim of nominating novel drug candidates for evaluation. These approaches, however, are prone to numerous limitations and potential biases that we have sought to address in the Drug Repurposing for Effective Alzheimer's Medicines (DREAM) study, a multi-step framework for selection and validation of potential drug candidates that has demonstrated the promise of STAT3 inhibitors and re-evaluated evidence for other drug candidates, such as phosphodiesterase inhibitors. Taken together, drug repurposing holds significant promise for development of novel AD therapeutics, particularly as the pace of data generation and development of analytical methods continue to accelerate.
Collapse
Affiliation(s)
- Jackson A Roberts
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, Massachusetts General Brigham, Boston, MA, USA
| | - Vijay R Varma
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Attila Jones
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
16
|
Espay AJ, Herrup K, Imbimbo BP, Kepp KP, Daly T. Recalibrating the Risk-Benefit Profiles of Lecanemab and Donanemab: Scales, Immunoreactivity, and Changes in Amyloid-β42. J Alzheimers Dis 2024; 99:877-881. [PMID: 38701151 DOI: 10.3233/jad-240171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Three recent anti-amyloid-β antibody trials for Alzheimer's disease reported similar effect sizes, used non-reactive saline as placebo, and showed large numbers of adverse events including imaging anomalies (ARIA) that correlate with cognitive changes. Conversely, all previous antibody trials were less reactive and pronounced ineffective. We argue that these observations point to unblinding bias, inflating apparent efficacy and thus altering the risk-benefit balance. Further, we highlight data demonstrating that beyond reducing amyloid, monoclonal antibodies increase monomeric amyloid-β42 in cerebrospinal fluid, which may explain potential benefits. We should recalibrate the efficacy of these antibodies and devote more resources into strategies beyond removing amyloid.
Collapse
Affiliation(s)
- Alberto J Espay
- Department of Neurology, James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, University of Cincinnati, Cincinnati, OH, USA
| | - Karl Herrup
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Kasper P Kepp
- Section of Biophysical and Biomedicinal Chemistry, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Timothy Daly
- Science Norms Democracy, UMR 8011 Sorbonne University, Paris, France
- Bioethics Program, FLACSO Argentina, Buenos Aires, Argentina
| |
Collapse
|
17
|
Kozin SA, Kechko OI, Adzhubei AA, Makarov AA, Mitkevich VA. Switching On/Off Amyloid Plaque Formation in Transgenic Animal Models of Alzheimer's Disease. Int J Mol Sci 2023; 25:72. [PMID: 38203242 PMCID: PMC10778642 DOI: 10.3390/ijms25010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
A hallmark of Alzheimer's disease (AD) are the proteinaceous aggregates formed by the amyloid-beta peptide (Aβ) that is deposited inside the brain as amyloid plaques. The accumulation of aggregated Aβ may initiate or enhance pathologic processes in AD. According to the amyloid hypothesis, any agent that has the capability to inhibit Aβ aggregation and/or destroy amyloid plaques represents a potential disease-modifying drug. In 2023, a humanized IgG1 monoclonal antibody (lecanemab) against the Aβ-soluble protofibrils was approved by the US FDA for AD therapy, thus providing compelling support to the amyloid hypothesis. To acquire a deeper insight on the in vivo Aβ aggregation, various animal models, including aged herbivores and carnivores, non-human primates, transgenic rodents, fish and worms were widely exploited. This review is based on the recent data obtained using transgenic animal AD models and presents experimental verification of the critical role in Aβ aggregation seeding of the interactions between zinc ions, Aβ with the isomerized Asp7 (isoD7-Aβ) and the α4β2 nicotinic acetylcholine receptor.
Collapse
Affiliation(s)
- Sergey A. Kozin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.I.K.); (A.A.A.); (A.A.M.)
| | | | | | | | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.I.K.); (A.A.A.); (A.A.M.)
| |
Collapse
|
18
|
Liu KY, Walsh S, Brayne C, Merrick R, Richard E, Howard R. Evaluation of clinical benefits of treatments for Alzheimer's disease. THE LANCET. HEALTHY LONGEVITY 2023; 4:e645-e651. [PMID: 37924845 DOI: 10.1016/s2666-7568(23)00193-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 11/06/2023] Open
Abstract
The need for regulatory approval of new therapies for the treatment of Alzheimer's disease-a progressive neurodegenerative condition-has made the assessment of treatment efficacy an urgent priority for discussion and investigation in the field. In the first part of this Personal View, we summarise current views on what constitutes a clinically meaningful benefit from treatment for Alzheimer's disease, including the concept of a minimum treatment effect against which to compare trial outcomes and its limitations. Considering existing and divergent definitions of clinically meaningful change, we define this concept in the second part of the Personal View by proposing a new approach that consecutively considers whether a treatment benefit for Alzheimer's disease is noticeable, valuable, and worthwhile in the context of costs and risks. This approach could be a useful foundation from which the field can move forwards on this issue and address existing gaps in understanding.
Collapse
Affiliation(s)
- Kathy Y Liu
- Division of Psychiatry, University College London, London, UK.
| | - Sebastian Walsh
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Carol Brayne
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Richard Merrick
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Edo Richard
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands; Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
19
|
Salemme S, Ancidoni A, Vanacore N. Responder analyses for anti-amyloid immunotherapies for Alzheimer's disease: a paradigm shift by regulatory authorities is urgently needed. Brain Commun 2023; 5:fcad276. [PMID: 37901042 PMCID: PMC10612473 DOI: 10.1093/braincomms/fcad276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 08/31/2023] [Accepted: 10/20/2023] [Indexed: 10/31/2023] Open
Affiliation(s)
- Simone Salemme
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena 41125, Italy
| | - Antonio Ancidoni
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome 00161, Italy
- Department of Public Health and Infectious Diseases, Sapienza University, Rome 00185, Italy
| | - Nicola Vanacore
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome 00161, Italy
| |
Collapse
|
20
|
Greenberg BD, Lemere CA, Barnes LL, Hayden KM, Kukull WA, Oh ES, Snyder PJ, Supiano M, Dilworth‐Anderson P. Prescribing anti-amyloid immunotherapies to treat Alzheimer's disease: Fully informing patient decisions. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2023; 9:e12426. [PMID: 37799322 PMCID: PMC10549961 DOI: 10.1002/trc2.12426] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 10/07/2023]
Affiliation(s)
- Barry D. Greenberg
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Cynthia A. Lemere
- Ann Romney Center for Neurologic DiseasesBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Lisa L. Barnes
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | | | - Walter A. Kukull
- Department of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Esther S. Oh
- Departments of MedicinePsychiatry and Behavioral Sciencesand PathologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Peter J. Snyder
- Department of Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Department of NeurologyAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Mark Supiano
- Division of Geriatrics, University of Utah Spencer Eccles Fox School of MedicineUniversity of Utah School on AgingSalt Lake CityUtahUSA
| | - Peggye Dilworth‐Anderson
- Gillings School of Global Public HealthUniversity of North Carolina‐Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
21
|
Cutuli D, Decandia D, Giacovazzo G, Coccurello R. Physical Exercise as Disease-Modifying Alternative against Alzheimer's Disease: A Gut-Muscle-Brain Partnership. Int J Mol Sci 2023; 24:14686. [PMID: 37834132 PMCID: PMC10572207 DOI: 10.3390/ijms241914686] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Alzheimer's disease (AD) is a common cause of dementia characterized by neurodegenerative dysregulations, cognitive impairments, and neuropsychiatric symptoms. Physical exercise (PE) has emerged as a powerful tool for reducing chronic inflammation, improving overall health, and preventing cognitive decline. The connection between the immune system, gut microbiota (GM), and neuroinflammation highlights the role of the gut-brain axis in maintaining brain health and preventing neurodegenerative diseases. Neglected so far, PE has beneficial effects on microbial composition and diversity, thus providing the potential to alleviate neurological symptoms. There is bidirectional communication between the gut and muscle, with GM diversity modulation and short-chain fatty acid (SCFA) production affecting muscle metabolism and preservation, and muscle activity/exercise in turn inducing significant changes in GM composition, functionality, diversity, and SCFA production. This gut-muscle and muscle-gut interplay can then modulate cognition. For instance, irisin, an exercise-induced myokine, promotes neuroplasticity and cognitive function through BDNF signaling. Irisin and muscle-generated BDNF may mediate the positive effects of physical activity against some aspects of AD pathophysiology through the interaction of exercise with the gut microbial ecosystem, neural plasticity, anti-inflammatory signaling pathways, and neurogenesis. Understanding gut-muscle-brain interconnections hold promise for developing strategies to promote brain health, fight age-associated cognitive decline, and improve muscle health and longevity.
Collapse
Affiliation(s)
- Debora Cutuli
- Department of Psychology, University of Rome La Sapienza, 00185 Rome, Italy;
- European Center for Brain Research, Santa Lucia Foundation IRCCS, 00143 Rome, Italy;
| | - Davide Decandia
- Department of Psychology, University of Rome La Sapienza, 00185 Rome, Italy;
- European Center for Brain Research, Santa Lucia Foundation IRCCS, 00143 Rome, Italy;
| | - Giacomo Giacovazzo
- European Center for Brain Research, Santa Lucia Foundation IRCCS, 00143 Rome, Italy;
- Facoltà di Medicina Veterinaria, Università degli Studi di Teramo (UniTE), 64100 Teramo, Italy
| | - Roberto Coccurello
- European Center for Brain Research, Santa Lucia Foundation IRCCS, 00143 Rome, Italy;
- Institute for Complex Systems (ISC), National Council of Research (CNR), 00185 Rome, Italy
| |
Collapse
|
22
|
Hardy J. Anti-amyloid therapies work for Alzheimer's disease. Brain Commun 2023; 5:fcad204. [PMID: 37492486 PMCID: PMC10365829 DOI: 10.1093/braincomms/fcad204] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/08/2023] [Accepted: 07/12/2023] [Indexed: 07/27/2023] Open
Affiliation(s)
- John Hardy
- Correspondence to: John Hardy, Reta Lilla Weston Research Laboratories and Department of Neurodegenerative Disease and Dementia Research Institute, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK E-mail:
| |
Collapse
|
23
|
Castellani RJ, Perry G. The Teflon hypothesis. Brain Commun 2023; 5:fcad203. [PMID: 37492487 PMCID: PMC10365830 DOI: 10.1093/braincomms/fcad203] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 06/29/2023] [Accepted: 07/12/2023] [Indexed: 07/27/2023] Open
Abstract
This scientific commentary refers to 'Key questions for the evaluation of anti-amyloid immunotherapies for Alzheimer's disease', by Liu et al. (https://doi.org/10.1093/braincomms/fcad175).
Collapse
Affiliation(s)
- Rudy J Castellani
- Correspondence to: Rudy J. Castellani, MD, Department of Pathology, Northwestern University Feinberg School of Medicine, 710 N. Fairbanks Court, Olson Pavilion 2-514, Chicago, IL 60611, USA E-mail:
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| |
Collapse
|
24
|
Salemme S, Ancidoni A, Locuratolo N, Piscopo P, Lacorte E, Canevelli M, Vanacore N. Advances in amyloid-targeting monoclonal antibodies for Alzheimer's disease: clinical and public health issues. Expert Rev Neurother 2023; 23:1113-1129. [PMID: 37975226 DOI: 10.1080/14737175.2023.2284305] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a major global public health challenge. To date, no treatments have been shown to stop the underlying pathological processes. The cerebral accumulation of amyloid-beta (Ab) is still considered as the primum movens of AD and disease-modifying treatments targeting Ab are reaching - or have already reached - clinical practice. AREAS COVERED The authors explore the main advancements from Aβ-targeting monoclonal antibodies (mAbs) for the treatment of AD. From a public health perspective, they address ethically relevant issues such as the benevolence and non-maleficence principles. They report on the potential biological and clinical benefits of these drugs, discussing minimal clinically important differences (MCID) and other relevant outcomes. They examine the short- and long-term effects of amyloid-related imaging abnormalities (ARIA), and explore the differences between eligibility criteria in clinical trials, appropriate use recommendations, and prescribing information content. In doing so, they contextualize the discussion on the disagreements among different regulatory authorities. EXPERT OPINION Although anti-β-amyloid monoclonal antibodies may be effective in selected scenarios, non-negligible knowledge gaps and implementation limits persist. Overcoming these gaps can no longer be postponed if we are to ensure the principles of Quality of Care for patients with cognitive impairment who would be eligible for this class of drugs.
Collapse
Affiliation(s)
- Simone Salemme
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonio Ancidoni
- National Centre for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Nicoletta Locuratolo
- National Centre for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| | - Paola Piscopo
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Eleonora Lacorte
- National Centre for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| | - Marco Canevelli
- National Centre for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
- Department of Human Neuroscience, "Sapienza" University, Rome, Italy
| | - Nicola Vanacore
- National Centre for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| |
Collapse
|