1
|
Pascoal TA, Aguzzoli CS, Lussier FZ, Crivelli L, Suemoto CK, Fortea J, Rosa-Neto P, Zimmer ER, Ferreira PCL, Bellaver B. Insights into the use of biomarkers in clinical trials in Alzheimer's disease. EBioMedicine 2024; 108:105322. [PMID: 39366844 DOI: 10.1016/j.ebiom.2024.105322] [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: 02/26/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 10/06/2024] Open
Abstract
Biomarkers have been instrumental in population selection and disease monitoring in clinical trials of recently FDA-approved drugs targeting amyloid-β to slow the progression of Alzheimer's disease (AD). As new therapeutic strategies and biomarker techniques emerge, the importance of biomarkers in drug development is growing exponentially. In this emerging landscape, biomarkers are expected to serve a wide range of contexts of use in clinical trials focusing on AD and related dementias. The joint FDA-NIH BEST (Biomarkers, EndpointS, and other Tools) framework provides standardised terminology to facilitate communication among stakeholders in this increasingly complex field. This review explores various applications of biomarkers relevant to AD clinical trials, using the BEST resource as a reference. For simplicity, we predominantly provide contextual characterizations of biomarkers use from the perspective of drugs targeting amyloid-β and tau proteins. However, general definitions and concepts can be extrapolated to other targets.
Collapse
Affiliation(s)
- Tharick A Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | | | - Firoza Z Lussier
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Lucía Crivelli
- Department of Cognitive Neurology, Fleni, Buenos Aires, Argentina
| | - Claudia K Suemoto
- Division of Geriatrics, University of São Paulo Medical School, São Paulo, Brazil
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
| | - Eduardo R Zimmer
- Brain Institute of Rio Grande do Sul, PUCRS, Porto Alegre, Brazil; Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil; Graduate Program in Biological Sciences, Biochemistry (PPGBioq), and Pharmacology and Therapeutics (PPGFT), Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Pamela C L Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
2
|
Schöll M, Verberk IMW, Del Campo M, Delaby C, Therriault J, Chong JR, Palmqvist S, Alcolea D. Challenges in the practical implementation of blood biomarkers for Alzheimer's disease. THE LANCET. HEALTHY LONGEVITY 2024; 5:100630. [PMID: 39369727 DOI: 10.1016/j.lanhl.2024.07.013] [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: 12/22/2023] [Revised: 06/23/2024] [Accepted: 07/29/2024] [Indexed: 10/08/2024] Open
Abstract
Blood biomarkers have emerged as accessible, cost-effective, and highly promising tools for advancing the diagnostics of Alzheimer's disease. However, transitioning from cerebrospinal fluid biomarkers to blood biomarkers-eg, to verify amyloid β pathology-requires careful consideration. This Series paper highlights the main challenges in the implementation of blood biomarkers for Alzheimer's disease in different possible contexts of use. Despite the robustness of measuring blood biomarker concentrations, the widespread adoption of blood biomarkers requires rigorous standardisation efforts to address inherent challenges in diverse contexts of use. The challenges include understanding the effect of pre-analytical and analytical conditions, potential confounding factors, and comorbidities that could influence outcomes of blood biomarkers and their use in diverse populations. Additionally, distinct scenarios present their own specific challenges. In memory clinics, the successful integration of blood biomarkers in diagnostic tests will require well-established diagnostic accuracy and comprehensive assessments of the effect of blood biomarkers on the diagnostic confidence and patient management of clinicians. In primary care settings, and even more when implemented in population-based screening programmes for which no experience with any biomarkers for Alzheimer's disease currently exists, the implementation of blood biomarkers will be challenged by the need for education of primary care clinical staff and clear guidelines. However, despite the challenges, blood biomarkers hold great promise for substantially enhancing the diagnostic accuracy and effectively streamlining referral processes, leading to earlier diagnosis and access to treatments. The ongoing efforts that are shaping the integration of blood biomarkers across diverse clinical settings pave the way towards precision medicine in Alzheimer's disease.
Collapse
Affiliation(s)
- Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Mölndal, Sweden; Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden; Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK; Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Marta Del Campo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute (IMIM), Barcelona, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Constance Delaby
- LBPC-PPC, University of Montpellier, CHU Montpellier, INM INSERM, Montpellier, France; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada; Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Memory, Aging and Cognition Centre, National University Health Systems, Singapore
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Clinical Sciences in Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
3
|
Ankeny SE, Bacci JR, Decourt B, Sabbagh MN, Mielke MM. Navigating the Landscape of Plasma Biomarkers in Alzheimer's Disease: Focus on Past, Present, and Future Clinical Applications. Neurol Ther 2024:10.1007/s40120-024-00658-x. [PMID: 39244522 DOI: 10.1007/s40120-024-00658-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/20/2024] [Indexed: 09/09/2024] Open
Abstract
As the prevalence of Alzheimer's disease (AD) and its impact on healthcare systems increase, developing tools for accurate diagnosis and monitoring of disease progression is a priority. Recent technological advancements have allowed for the development of blood-based biomarkers (BBMs) to aid in the diagnosis of AD, but many questions remain regarding the clinical implementation of these BBMs. This review outlines the historical timeline of AD BBM development. It highlights key breakthroughs that have transformed the perspective of AD BBMs from theoretically ideal but unattainable markers, to clinically valid and reliable BBMs with potential for implementation in healthcare settings. Technological advancements like single-molecule detection and mass spectrometry methods have significantly improved assay sensitivity and accuracy. High-throughput, fully automated platforms have potential for clinical use. Despite these advancements, however, significant work is needed before AD BBMs can be implemented in widespread clinical practice. Cutpoints must be established, the influence of chronic conditions and medications on BBM levels must be better understood, and guidelines must be created for healthcare providers related to interpreting and communicating information obtained from AD BBMs. Additionally, the development of BBMs for synaptic dysfunction, inflammation, and cerebrovascular disease may provide better precision medicine approaches to treating AD and related dementia. Future research and collaboration between scientists and physicians are essential to addressing these challenges and further advancing AD BBMs, with the goal of integration in clinical practice.
Collapse
Affiliation(s)
- Sarrah E Ankeny
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Julia R Bacci
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Boris Decourt
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Marwan N Sabbagh
- Alzheimer's and Memory Disorders Division, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
4
|
Delvenne A, Gobom J, Schindler SE, Kate MT, Reus LM, Dobricic V, Tijms BM, Benzinger TLS, Cruchaga C, Teunissen CE, Ramakers I, Martinez‐Lage P, Tainta M, Vandenberghe R, Schaeverbeke J, Engelborghs S, Roeck ED, Popp J, Peyratout G, Tsolaki M, Freund‐Levi Y, Lovestone S, Streffer J, Barkhof F, Bertram L, Blennow K, Zetterberg H, Visser PJ, Vos SJB. CSF proteomic profiles of neurodegeneration biomarkers in Alzheimer's disease. Alzheimers Dement 2024; 20:6205-6220. [PMID: 38970402 PMCID: PMC11497678 DOI: 10.1002/alz.14103] [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: 04/03/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/08/2024]
Abstract
INTRODUCTION We aimed to unravel the underlying pathophysiology of the neurodegeneration (N) markers neurogranin (Ng), neurofilament light (NfL), and hippocampal volume (HCV), in Alzheimer's disease (AD) using cerebrospinal fluid (CSF) proteomics. METHODS Individuals without dementia were classified as A+ (CSF amyloid beta [Aβ]42), T+ (CSF phosphorylated tau181), and N+ or N- based on Ng, NfL, or HCV separately. CSF proteomics were generated and compared between groups using analysis of covariance. RESULTS Only a few individuals were A+T+Ng-. A+T+Ng+ and A+T+NfL+ showed different proteomic profiles compared to A+T+Ng- and A+T+NfL-, respectively. Both Ng+ and NfL+ were associated with neuroplasticity, though in opposite directions. Compared to A+T+HCV-, A+T+HCV+ showed few proteomic changes, associated with oxidative stress. DISCUSSION Different N markers are associated with distinct neurodegenerative processes and should not be equated. N markers may differentially complement disease staging beyond amyloid and tau. Our findings suggest that Ng may not be an optimal N marker, given its low incongruency with tau pathophysiology. HIGHLIGHTS In Alzheimer's disease, neurogranin (Ng)+, neurofilament light (NfL)+, and hippocampal volume (HCV)+ showed differential protein expression in cerebrospinal fluid. Ng+ and NfL+ were associated with neuroplasticity, although in opposite directions. HCV+ showed few proteomic changes, related to oxidative stress. Neurodegeneration (N) markers may differentially refine disease staging beyond amyloid and tau. Ng might not be an optimal N marker, as it relates more closely to tau.
Collapse
|
5
|
Strobel J, Yousefzadeh-Nowshahr E, Deininger K, Bohn KP, von Arnim CAF, Otto M, Solbach C, Anderl-Straub S, Polivka D, Fissler P, Glatting G, Riepe MW, Higuchi M, Beer AJ, Ludolph A, Winter G. Exploratory Tau PET/CT with [11C]PBB3 in Patients with Suspected Alzheimer's Disease and Frontotemporal Lobar Degeneration: A Pilot Study on Correlation with PET Imaging and Cerebrospinal Fluid Biomarkers. Biomedicines 2024; 12:1460. [PMID: 39062033 PMCID: PMC11274645 DOI: 10.3390/biomedicines12071460] [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: 05/08/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Accurately diagnosing Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD) is challenging due to overlapping symptoms and limitations of current imaging methods. This study investigates the use of [11C]PBB3 PET/CT imaging to visualize tau pathology and improve diagnostic accuracy. Given diagnostic challenges with symptoms and conventional imaging, [11C]PBB3 PET/CT's potential to enhance accuracy was investigated by correlating tau pathology with cerebrospinal fluid (CSF) biomarkers, positron emission tomography (PET), computed tomography (CT), amyloid-beta, and Mini-Mental State Examination (MMSE). We conducted [11C]PBB3 PET/CT imaging on 24 patients with suspected AD or FTLD, alongside [11C]PiB PET/CT (13 patients) and [18F]FDG PET/CT (15 patients). Visual and quantitative assessments of [11C]PBB3 uptake using standardized uptake value ratios (SUV-Rs) and correlation analyses with clinical assessments were performed. The scans revealed distinct tau accumulation patterns; 13 patients had no or faint uptake (PBB3-negative) and 11 had moderate to pronounced uptake (PBB3-positive). Significant inverse correlations were found between [11C]PBB3 SUV-Rs and MMSE scores, but not with CSF-tau or CSF-amyloid-beta levels. Here, we show that [11C]PBB3 PET/CT imaging can reveal distinct tau accumulation patterns and correlate these with cognitive impairment in neurodegenerative diseases. Our study demonstrates the potential of [11C]PBB3-PET imaging for visualizing tau pathology and assessing disease severity, offering a promising tool for enhancing diagnostic accuracy in AD and FTLD. Further research is essential to validate these findings and refine the use of tau-specific PET imaging in clinical practice, ultimately improving patient care and treatment outcomes.
Collapse
Affiliation(s)
- Joachim Strobel
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Katharina Deininger
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Karl Peter Bohn
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Markus Otto
- Department of Neurology, Halle University, 06120 Halle, Germany
| | - Christoph Solbach
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Dörte Polivka
- Department of Neurology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Patrick Fissler
- Psychiatric Services Thurgau (Academic Teaching Hospital of the University of Konstanz), 8596 Münsterlingen, Switzerland
| | - Gerhard Glatting
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Matthias W. Riepe
- Department of Psychiatry and Psychotherapy II, Ulm University, 89075 Ulm, Germany
| | - Makoto Higuchi
- National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Ambros J. Beer
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Albert Ludolph
- Department of Neurology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Gordon Winter
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
6
|
Reiss AB, Pinkhasov A. Special Issue "Commemorative Issue Celebrating the 20th Anniversary of the Alzheimer's Foundation of America: Understanding and Treating Alzheimer's Disease". MEDICINA (KAUNAS, LITHUANIA) 2024; 60:712. [PMID: 38792895 PMCID: PMC11122752 DOI: 10.3390/medicina60050712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in older persons [...].
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | | |
Collapse
|
7
|
Silva-Spínola A, Leitão MJ, Nadal A, Le Bastard N, Santana I, Baldeiras I. Exploring the potential of fully automated LUMIPULSE G plasma assays for detecting Alzheimer's disease pathology. Alzheimers Res Ther 2024; 16:51. [PMID: 38454502 PMCID: PMC10918996 DOI: 10.1186/s13195-024-01397-9] [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: 07/25/2023] [Accepted: 01/26/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND LUMIPULSE G-automated immunoassays represent a widely used method for the quantification of Alzheimer's disease (AD) biomarkers in the cerebrospinal fluid (CSF). Less invasive blood-based markers confer a promising tool for AD diagnosis at prodromal stages (mild cognitive impairment (MCI)). Highly sensitive assays for the quantification of amyloid-beta (Aβ) and phosphorylated Tau-181 (p-Tau181) in the blood are showing promising results. In this study, we evaluated the clinical performance of the recently available fully automated LUMIPULSE plasma marker assays for detecting brain AD pathology and for predicting progression from MCI to AD dementia stage. METHODS A retrospective exploratory cohort of 138 individuals (22 neurological controls [NC], 72 MCI, and 44 AD dementia patients) was included. Data regarding baseline CSF concentrations of Aβ42, Aβ40, t-Tau, and p-Tau181 was available and used to establish the presence of AD brain pathology. Baseline Aβ42, Aβ40, and p-Tau181 concentrations were determined in stored plasma samples using high-throughput fully automated LUMIPULSE assays. Progression from MCI to AD dementia was evaluated during follow-up (mean 6.4 ± 2.5 years). Moreover, a prospective validation cohort of 72 individuals with memory complaints underwent AD biomarker quantification, closely mirroring typical clinical practice. This cohort aimed to confirm the study's main findings. RESULTS In the exploratory cohort, correlations between CSF and plasma were moderate for p-Tau181 (ρ = 0.61, p < 0.001) and weak for Aβ42/Aβ40 ratio (ρ = 0.39, p < 0.001). Plasma p-Tau181 and p-Tau181/Aβ42 concentrations were significantly increased while Aβ42/Aβ40 was significantly decreased (p < 0.001) in patients with AD dementia and prodromal AD, as well as in individuals with CSF abnormal amyloid concentrations (A +). Plasma p-Tau181 showed a robust performance in differentiating patients clinically diagnosed as AD (AUC = 0.89; 95% CI 0.83-0.94); A + vs. A - (AUC = 0.84, 95% CI 0.77-0.91) and also in predicting conversion to AD dementia in MCI patients (AUC = 0.89, 95% CI 0.81-0.96). When tested in the validation cohort, plasma p-Tau181 displayed 83.3% of the overall percentage of agreement according to amyloid status. CONCLUSIONS Our results show that the measurement of p-Tau181 in plasma has great potential as a non-invasive prognostic screening tool for implementation in a clinical setting.
Collapse
Affiliation(s)
- Anuschka Silva-Spínola
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- Centre for Informatics and Systems, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal.
- Neurochemistry Laboratory, Neurology Department, Coimbra University Hospital, Praceta Mota Pinto, 3004-561, Coimbra, Portugal.
| | - Maria João Leitão
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Neurochemistry Laboratory, Neurology Department, Coimbra University Hospital, Praceta Mota Pinto, 3004-561, Coimbra, Portugal
| | | | | | - Isabel Santana
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar E Universitário de Coimbra, Coimbra, Portugal
| | - Inês Baldeiras
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Neurochemistry Laboratory, Neurology Department, Coimbra University Hospital, Praceta Mota Pinto, 3004-561, Coimbra, Portugal
| |
Collapse
|
8
|
Elghanam Y, Purja S, Kim EY. Biomarkers as Endpoints in Clinical Trials for Alzheimer's Disease. J Alzheimers Dis 2024; 99:693-703. [PMID: 38669547 DOI: 10.3233/jad-240008] [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: 04/28/2024]
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease that imposes economic and societal burden. Biomarkers have played a crucial role in the recent approval of aducanumab and lecanemab as disease-modifying therapies which marked a significant milestone for the treatment of AD. The inclusion of biomarkers in AD trials facilitates precise diagnosis, monitors safety, demonstrates target engagement, and supports disease modification. Objective This study analyzed the utilization state and trends of biomarkers as endpoints in AD trials. Methods In this retrospective study, trials were collected by searching clinicaltrials.gov using the term "Alzheimer". Primary and secondary outcomes were analyzed separately for each phase. Results Among the 1,048 analyzed trials, 313 (29.87%) adopted biomarkers as primary endpoints and 364 (34.73%) as secondary endpoints, mainly in phases 1 and 2. The top three biomarkers adopted as primary endpoints in phases 1, 2, and 3 were amyloid-PET, tau-PET, and MRI. The top three biomarkers adopted as secondary endpoints, in phase 1, were cerebrospinal fluid (CSF) amyloid-β (Aβ), blood Aβ and amyloid-PET; in phase 2, they were MRI, CSF Aβ, and CSF phospho-tau; and in phase 3, they were amyloid PET, MRI, and blood Aβ. There was a statistically significant increase in the adoption of biomarkers as primary endpoints in phase 2 trials (p = 0.001) and secondary endpoints in phase 3 trials (p = 0.001). Conclusions The growing recognition of the importance of biomarkers in AD trial' design and drug development is evident by the significant steady increase in biomarkers' utilization in phases 2 and 3.
Collapse
Affiliation(s)
- Yomna Elghanam
- Department of Health, Evidence-Based and Clinical Research Laboratory, Social, and Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Sujata Purja
- Department of Health, Evidence-Based and Clinical Research Laboratory, Social, and Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Eun Young Kim
- Department of Health, Evidence-Based and Clinical Research Laboratory, Social, and Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul, Korea
- The Graduate School for Pharmaceutical Industry Management, College of Pharmacy, Chung-Ang University, Seoul, Korea
- The Department of Pharmaceutical Regulatory Sciences, Chung-Ang University, Seoul, Korea
| |
Collapse
|
9
|
Schicktanz S, Alpinar-Segawa Z, Ulitsa N, Perry J, Werner P. Moving Towards Ethical-Practical Recommendations for Alzheimer's Disease Prediction: Addressing Interindividual, Interprofessional, and Societal Aspects. J Alzheimers Dis 2024; 101:1063-1081. [PMID: 39269830 DOI: 10.3233/jad-231137] [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: 09/15/2024]
Abstract
Biomarkers for predicting Alzheimer's disease (AD) are advancing and their implementation in various healthcare systems is imminent. There is a need for ethical standards addressing information needs, socio-ethical concerns, and expectations of healthy and at-risk persons. We present an ethical approach that integrates different existing ethical frameworks and discussion of our empirical, cross-cultural findings in a multi-layered perspective by addressing three levels. The micro-level focuses on the communication between counseling professionals, persons at risk or in an early stage of dementia, and family members. The meso-level addresses interprofessional cooperation and exchange as a key element for best person-centered care. The macro-level considers public health promotion, the media, and public-funded research. This approach allows to address key ethical concepts including beneficence, non-maleficence, autonomy, informational self-determination, empowerment, and justice. Our contribution specifically examines the ethical challenges associated with AD prediction by means of biomarkers, based on insights from a German-Israeli comparison, and promotes a transdisciplinary discussion across different healthcare contexts. We propose a reflection on three levels to go beyond the clinical counseling context and to consider the rapidly evolving field of biomarkers in the coming years. Our ethical-practical recommendations should not be considered final, but rather procedural and will require continuous adaptation regarding culturally varying practices, new algorithms, meta-analyses, and re-evaluation of established recommendations.
Collapse
Affiliation(s)
- Silke Schicktanz
- Department of Medical Ethics and History of Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Zümrüt Alpinar-Segawa
- Department of Medical Ethics and History of Medicine, University Medical Center Göttingen, Göttingen, Germany
- Institute for Ethics, History and Philosophy of Medicine, Hannover Medical School, Hannover, Germany
| | - Natalie Ulitsa
- Department of Medical Ethics and History of Medicine, University Medical Center Göttingen, Göttingen, Germany
- Department of Community Mental Health, Faculty of Social Welfare & Health Sciences, University of Haifa, Haifa, Israel
| | - Julia Perry
- Department of Medical Ethics and History of Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Perla Werner
- Department of Community Mental Health, Faculty of Social Welfare & Health Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
10
|
Brown JA, Lee AJ, Fernhoff K, Pistone T, Pasquini L, Wise AB, Staffaroni AM, Luisa Mandelli M, Lee SE, Boxer AL, Rankin KP, Rabinovici GD, Luisa Gorno Tempini M, Rosen HJ, Kramer JH, Miller BL, Seeley WW. Functional network collapse in neurodegenerative disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569654. [PMID: 38106054 PMCID: PMC10723363 DOI: 10.1101/2023.12.01.569654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Cognitive and behavioral deficits in Alzheimer's disease (AD) and frontotemporal dementia (FTD) result from brain atrophy and altered functional connectivity. However, it is unclear how atrophy relates to functional connectivity disruptions across dementia subtypes and stages. We addressed this question using structural and functional MRI from 221 patients with AD (n=82), behavioral variant FTD (n=41), corticobasal syndrome (n=27), nonfluent (n=34) and semantic (n=37) variant primary progressive aphasia, and 100 cognitively normal individuals. Using partial least squares regression, we identified three principal structure-function components. The first component showed overall atrophy correlating with primary cortical hypo-connectivity and subcortical/association cortical hyper-connectivity. Components two and three linked focal syndrome-specific atrophy to peri-lesional hypo-connectivity and distal hyper-connectivity. Structural and functional component scores predicted global and domain-specific cognitive deficits. Anatomically, functional connectivity changes reflected alterations in specific brain activity gradients. Eigenmode analysis identified temporal phase and amplitude collapse as an explanation for atrophy-driven functional connectivity changes.
Collapse
Affiliation(s)
- Jesse A. Brown
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Alex J. Lee
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Kristen Fernhoff
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Taylor Pistone
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Lorenzo Pasquini
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Amy B. Wise
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Adam M. Staffaroni
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Maria Luisa Mandelli
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Suzee E. Lee
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Adam L. Boxer
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Katherine P. Rankin
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Gil D. Rabinovici
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Maria Luisa Gorno Tempini
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Howard J. Rosen
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Joel H. Kramer
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - Bruce L. Miller
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | - William W. Seeley
- University of California, San Francisco, Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, San Francisco, CA, USA
| | | |
Collapse
|
11
|
Lalwani RC, Volmar CH, Wahlestedt C, Webster KA, Shehadeh LA. Contextualizing the Role of Osteopontin in the Inflammatory Responses of Alzheimer's Disease. Biomedicines 2023; 11:3232. [PMID: 38137453 PMCID: PMC10741223 DOI: 10.3390/biomedicines11123232] [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: 10/25/2023] [Revised: 12/01/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive accumulations of extracellular amyloid-beta (Aβ) aggregates from soluble oligomers to insoluble plaques and hyperphosphorylated intraneuronal tau, also from soluble oligomers to insoluble neurofibrillary tangles (NFTs). Tau and Aβ complexes spread from the entorhinal cortex of the brain to interconnected regions, where they bind pattern recognition receptors on microglia and astroglia to trigger inflammation and neurotoxicity that ultimately lead to neurodegeneration and clinical AD. Systemic inflammation is initiated by Aβ's egress into the circulation, which may be secondary to microglial activation and can confer both destructive and reparative actions. Microglial activation pathways and downstream drivers of Aβ/NFT neurotoxicity, including inflammatory regulators, are primary targets for AD therapy. Osteopontin (OPN), an inflammatory cytokine and biomarker of AD, is implicated in Aβ clearance and toxicity, microglial activation, and inflammation, and is considered to be a potential therapeutic target. Here, using the most relevant works from the literature, we review and contextualize the evidence for a central role of OPN and associated inflammation in AD.
Collapse
Affiliation(s)
- Roshni C. Lalwani
- Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Claude-Henry Volmar
- Department of Psychiatry, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.-H.V.); (C.W.)
- Center for Therapeutic Innovation, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Claes Wahlestedt
- Department of Psychiatry, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.-H.V.); (C.W.)
- Center for Therapeutic Innovation, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Keith A. Webster
- Integene International Holdings, LLC, Miami, FL 33137, USA;
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
- Everglades BioPharma, Houston, TX 77098, USA
| | - Lina A. Shehadeh
- Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
12
|
Singh S, Mahajan M, Kumar D, Singh K, Chowdhary M, Amit. An inclusive study of recent advancements in Alzheimer's disease: A comprehensive review. Neuropeptides 2023; 102:102369. [PMID: 37611472 DOI: 10.1016/j.npep.2023.102369] [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: 06/17/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023]
Abstract
Alzheimer's disease (AD) has remained elusive in revealing its pathophysiology and mechanism of development. In this review paper, we attempt to highlight several theories that abound about the exact pathway of AD development. The number of cases worldwide has prompted a constant flow of research to detect high-risk patients, slow the progression of the disease and discover improved methods of treatment that may prove effective. We shall focus on the two main classes of drugs that are currently in use; and emerging ones with novel mechanisms that are under development. As of late there has also been increased attention towards factors that were previously thought to be unrelated to AD, such as the gut microbiome, lifestyle habits, and diet. Studies have now shown that all these factors make an impact on AD progression, thus bringing to our attention more areas that could hold the key to combating this disease. This paper covers all the aforementioned factors concisely. We also briefly explore the relationship between mental health and AD, both before and after the diagnosis of the disease.
Collapse
Affiliation(s)
- Sukanya Singh
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Mitali Mahajan
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Dhawal Kumar
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Kunika Singh
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Mehvish Chowdhary
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Amit
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India.
| |
Collapse
|
13
|
Blennow K, Galasko D, Perneczky R, Quevenco FC, van der Flier WM, Akinwonmi A, Carboni M, Jethwa A, Suridjan I, Zetterberg H. The potential clinical value of plasma biomarkers in Alzheimer's disease. Alzheimers Dement 2023; 19:5805-5816. [PMID: 37694991 DOI: 10.1002/alz.13455] [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: 05/22/2023] [Revised: 08/02/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Many people with cognitive complaints or impairment never receive an accurate diagnosis of the underlying condition, potentially impacting their access to appropriate treatment. To address this unmet need, plasma biomarker tests are being developed for use in Alzheimer's disease (AD). Plasma biomarker tests span various stages of development, including in vitro diagnostic devices (or tests) (IVDs), laboratory-developed tests (LDTs) and research use only devices (or tests) (RUOs). Understanding the differences between each test type is important for appropriate implementation into the AD diagnostic pathway and care continuum. METHODS Authors reviewed scientific literature (PubMed, meeting abstracts and presentations, company press releases and websites) on AD plasma biomarkers. RESULTS This article defines IVDs, LDTs, and RUOs, discusses potential clinical applications and highlights the steps necessary for their clinical implementation. DISCUSSION Plasma biomarkers could revolutionize many areas of the AD diagnostic pathway and care continuum, but further research is needed. HIGHLIGHTS There is a need for a minimally invasive Alzheimer's disease (AD) diagnostic tool. AD plasma biomarker tests exist at various stages of commercial development. Understanding the development stage of a test is important for its appropriate use. Plasma biomarker tests could function as a triage tool to streamline AD diagnosis. Further steps remain before AD plasma biomarkers can be used routinely.
Collapse
Affiliation(s)
- Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, University Hospital of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Broomhall, Sheffield, UK
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
| | | | - Wiesje M van der Flier
- Department of Neurology, Alzheimer Centre Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Epidemiology and Data Science, Amsterdam UMC, Amsterdam, The Netherlands
| | - Akin Akinwonmi
- Roche Diagnostics International Ltd, Rotkreuz, Switzerland
| | | | | | | | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute, UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Special Administrative Region, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
14
|
Cummings JL, Gonzalez MI, Pritchard MC, May PC, Toledo-Sherman LM, Harris GA. The therapeutic landscape of tauopathies: challenges and prospects. Alzheimers Res Ther 2023; 15:168. [PMID: 37803386 PMCID: PMC10557207 DOI: 10.1186/s13195-023-01321-7] [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: 05/26/2023] [Accepted: 09/28/2023] [Indexed: 10/08/2023]
Abstract
Tauopathies are a group of neurodegenerative disorders characterized by the aggregation of the microtubule-associated protein tau. Aggregates of misfolded tau protein are believed to be implicated in neuronal death, which leads to a range of symptoms including cognitive decline, behavioral change, dementia, and motor deficits. Currently, there are no effective treatments for tauopathies. There are four clinical candidates in phase III trials and 16 in phase II trials. While no effective treatments are currently approved, there is increasing evidence to suggest that various therapeutic approaches may slow the progression of tauopathies or improve symptoms. This review outlines the landscape of therapeutic drugs (indexed through February 28, 2023) that target tau pathology and describes drug candidates in clinical development as well as those in the discovery and preclinical phases. The review also contains information on notable therapeutic programs that are inactive or that have been discontinued from development.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas (UNLV), Henderson, NV, USA
| | | | | | - Patrick C May
- ADvantage Neuroscience Consulting LLC, Fort Wayne, IN, USA
| | | | - Glenn A Harris
- Rainwater Charitable Foundation, 777 Main Street, Suite 2250, Fort Worth, TX, 76102, USA.
| |
Collapse
|
15
|
Papay RS, Stauffer SR, Perez DM. A PAM of the α 1A-Adrenergic receptor rescues biomarker, long-term potentiation, and cognitive deficits in Alzheimer's disease mouse models without effects on blood pressure. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2023; 5:100160. [PMID: 37448695 PMCID: PMC10336260 DOI: 10.1016/j.crphar.2023.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/30/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
α1-Adrenergic Receptors (ARs) regulate the sympathetic nervous system by the binding of norepinephrine (NE) and epinephrine (Epi) through different subtypes (α1A, α1B, α1D). α1A-AR activation is hypothesized to be memory forming and cognitive enhancing but drug development has been stagnant due to unwanted side effects on blood pressure. We recently reported the pharmacological characterization of the first positive allosteric modulator (PAM) for the α1A-AR with predictive pro-cognitive and memory properties. In this report, we now demonstrate the in vivo characteristics of Compound 3 (Cmpd-3) in two genetically-different Alzheimer's Disease (AD) mouse models. Drug metabolism and pharmacokinetic studies indicate sufficient brain penetrance and rapid uptake into the brain with low to moderate clearance, and a favorable inhibition profile against the major cytochrome p450 enzymes. Oral administration of Cmpd-3 (3-9 mg/kg QD) can fully rescue long-term potentiation defects and AD biomarker profile (amyloid β-40, 42) within 3 months of dosing to levels that were non-significant from WT controls and which outperformed donepezil (1 mg/kg QD). There were also significant effects on paired pulse facilitation and cognitive behavior. Long-term and high-dose in vivo studies with Cmpd-3 revealed no effects on blood pressure. Our results suggest that Cmpd-3 can maintain lasting therapeutic levels and efficacy with disease modifying effects with a once per day dosing regimen in AD mouse models with no observed side effects.
Collapse
Affiliation(s)
- Robert S. Papay
- The Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| | - Shaun R. Stauffer
- Center of Therapeutics Discovery, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, Ohio, 44195, USA
| | - Dianne M. Perez
- The Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| |
Collapse
|
16
|
Yatham LN. Biomarkers for clinical use in psychiatry: where are we and will we ever get there? World Psychiatry 2023; 22:263-264. [PMID: 37159356 PMCID: PMC10168147 DOI: 10.1002/wps.21079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 05/11/2023] Open
Affiliation(s)
- Lakshmi N Yatham
- Department of Psychiatry, Institute of Mental Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
17
|
Kim J, Choi P, Park YT, Kim T, Ham J, Kim JC. The Cannabinoids, CBDA and THCA, Rescue Memory Deficits and Reduce Amyloid-Beta and Tau Pathology in an Alzheimer’s Disease-like Mouse Model. Int J Mol Sci 2023; 24:ijms24076827. [PMID: 37047798 PMCID: PMC10095267 DOI: 10.3390/ijms24076827] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/01/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023] Open
Abstract
Most studies related to hemp are focused on Cannabidiol (CBD) and Tetrahydrocannabinol (THC); however, up to 120 types of phytocannabinoids are present in hemp. Hemp leaves contain large amounts of Cannabidiolic acid (CBDA) and Tetrahydrocannabinolic acid (THCA), which are acidic variants of CBD and THC and account for the largest proportion of CBDA. In recent studies, CBDA exhibited anti-hyperalgesia and anti-inflammatory effects. THCA also showed anti-inflammatory and neuroprotective effects that may be beneficial for treating neurodegenerative diseases. CBDA and THCA can penetrate the blood–brain barrier (BBB) and affect the central nervous system. The purpose of this study was to determine whether CBDA and THCA ameliorate Alzheimer’s disease (AD)-like features in vitro and in vivo. The effect of CBDA and THCA was evaluated in the Aβ1–42-treated mouse model. We observed that Aβ1–42-treated mice had more hippocampal Aβ and p-tau levels, pathological markers of AD, and loss of cognitive function compared with PBS-treated mice. However, CBDA- and THCA-treated mice showed decreased hippocampal Aβ and p-tau and superior cognitive function compared with Aβ1–42-treated mice. In addition, CBDA and THCA lowered Aβ and p-tau levels, alleviated calcium dyshomeostasis, and exhibited neuroprotective effects in primary neurons. Our results suggest that CBDA and THCA have anti-AD effects and mitigate memory loss and resilience to increased hippocampal Ca2+, Aβ, and p-tau levels. Together, CBDA and THCA may be useful therapeutic agents for treating AD.
Collapse
Affiliation(s)
- Juyong Kim
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
| | - Pilju Choi
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
| | - Young-Tae Park
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
| | - Taejung Kim
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Jungyeob Ham
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
- NeoCannBio Co., Ltd., Gangneung 02792, Republic of Korea
| | - Jin-Chul Kim
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| |
Collapse
|
18
|
Cummings J, Zhou Y, Lee G, Zhong K, Fonseca J, Cheng F. Alzheimer's disease drug development pipeline: 2023. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2023; 9:e12385. [PMID: 37251912 PMCID: PMC10210334 DOI: 10.1002/trc2.12385] [Citation(s) in RCA: 133] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 05/31/2023]
Abstract
Introduction Drugs that prevent the onset, slow progression, or improve cognitive and behavioral symptoms of Alzheimer's disease (AD) are needed. Methods We searched ClinicalTrials.gov for all current Phase 1, 2 and 3 clinical trials for AD and mild cognitive impairment (MCI) attributed to AD. We created an automated computational database platform to search, archive, organize, and analyze the derived data. The Common Alzheimer's Disease Research Ontology (CADRO) was used to identify treatment targets and drug mechanisms. Results On the index date of January 1, 2023, there were 187 trials assessing 141 unique treatments for AD. Phase 3 included 36 agents in 55 trials; 87 agents were in 99 Phase 2 trials; and Phase 1 had 31 agents in 33 trials. Disease-modifying therapies were the most common drugs comprising 79% of drugs in trials. Twenty-eight percent of candidate therapies are repurposed agents. Populating all current Phase 1, 2, and 3 trials will require 57,465 participants. Discussion The AD drug development pipeline is advancing agents directed at a variety of target processes. HIGHLIGHTS There are currently 187 trials assessing 141 drugs for the treatment of Alzheimer's disease (AD).Drugs in the AD pipeline address a variety of pathological processes.More than 57,000 participants will be required to populate all currently registered trials.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Department of Brain HealthChambers‐Grundy Center for Transformative NeuroscienceSchool of Integrated Health SciencesUniversity of Nevada, Las Vegas (UNLV)Las VegasNevadaUSA
- Department of Computer ScienceHoward R. Hughes College of EngineeringUniversity of Nevada, Las Vegas (UNLV)Las VegasNevadaUSA
| | - Yadi Zhou
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Garam Lee
- Department of Brain HealthSchool of Integrated Health SciencesUniversity of Nevada, Las Vegas (UNLV)Las VegasNevadaUSA
| | - Kate Zhong
- Department of Brain HealthChambers‐Grundy Center for Transformative NeuroscienceSchool of Integrated Health SciencesUniversity of Nevada, Las Vegas (UNLV)Las VegasNevadaUSA
- Department of Computer ScienceHoward R. Hughes College of EngineeringUniversity of Nevada, Las Vegas (UNLV)Las VegasNevadaUSA
| | - Jorge Fonseca
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
| | - Feixiong Cheng
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Case Comprehensive Cancer CenterCase Western Reserve University School of MedicineClevelandOhioUSA
| |
Collapse
|
19
|
Zhang J, Campion S, Catlin N, Reagan WJ, Palyada K, Ramaiah SK, Ramanathan R. Circulating microRNAs as promising testicular translatable safety biomarkers: current state and future perspectives. Arch Toxicol 2023; 97:947-961. [PMID: 36795116 PMCID: PMC9933818 DOI: 10.1007/s00204-023-03460-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/07/2023] [Indexed: 02/17/2023]
Abstract
Drug-induced testicular injury (DITI) is one of the often-observed and challenging safety issues seen during drug development. Semen analysis and circulating hormones currently utilized have significant gaps in their ability to detect testicular damage accurately. In addition, no biomarkers enable a mechanistic understanding of the damage to the different regions of the testis, such as seminiferous tubules, Sertoli, and Leydig cells. MicroRNAs (miRNAs) are a class of non-coding RNAs that modulate gene expression post-transcriptionally and have been indicated to regulate a wide range of biological pathways. Circulating miRNAs can be measured in the body fluids due to tissue-specific cell injury/damage or toxicant exposure. Therefore, these circulating miRNAs have become attractive and promising non-invasive biomarkers for assessing drug-induced testicular injury, with several reports on their use as safety biomarkers for monitoring testicular damage in preclinical species. Leveraging emerging tools such as 'organs-on-chips' that can emulate the human organ's physiological environment and function is starting to enable biomarker discovery, validation, and clinical translation for regulatory qualification and implementation in drug development.
Collapse
Affiliation(s)
- Jiangwei Zhang
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 10777 Science Center Dr, San Diego, CA, USA
| | - Sarah Campion
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA
| | - Natasha Catlin
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA
| | - William J Reagan
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA
| | - Kiran Palyada
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 10777 Science Center Dr, San Diego, CA, USA
| | - Shashi K Ramaiah
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, 02139, USA
| | - Ragu Ramanathan
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA.
| |
Collapse
|
20
|
Cummings J, Leisgang Osse AM, Kinney J. Geroscience and Alzheimer's Disease Drug Development. J Prev Alzheimers Dis 2023; 10:620-632. [PMID: 37874083 PMCID: PMC10720397 DOI: 10.14283/jpad.2023.103] [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: 10/25/2023]
Abstract
Age is the most important risk factor for Alzheimer's disease (AD). The acceptable age range for participation in AD clinical trials is 50 to 90, and this 40-year span incorporates enormous age-related change. Clinical trial participants tend to be younger and healthier than the general population. They are also younger than the general population of AD patients. Drug development from a geroscience perspective would take greater account of effects of aging on clinical trial outcomes. The AD clinical trial pipeline has diversified beyond the canonical targets of amyloid beta protein and tau. Many of these interventions apply to age-related disorders. Anti-inflammatory agents and bioenergetic and metabolic therapies are among the well represented classes in the pipeline and are applicable to AD and non-AD age-related conditions. Drug development strategies can be adjusted to better inform outcomes of trials regarding aged individuals. Inclusion of older individuals in the multiple ascending dose trials of Phase 1, use of geriatric-related clinical outcomes and biomarkers in Phase 2, and extension of these Phase 2 learnings to Phase 3 will result in a more comprehensive understanding of AD therapies and their relationship to aging. Clinical trials can employ a more comprehensive geriatric assessment approach and biomarkers more relevant to aging at baseline and as exploratory outcomes. Greater attention to the role of aging and its influence in AD clinical trials can result in better understanding of the generalizability of clinical trial findings to the older AD population.
Collapse
Affiliation(s)
- J Cummings
- Jeffrey Cummings, 1380 Opal valley street, Henderson, Nevada 89052, USA,
| | | | | |
Collapse
|
21
|
Abstract
This review deals with an unwelcome reality about several forms of dementia, including Alzheimer's disease- that these dementias are caused, in part or whole, by the aging of the vasculature. Since the vasculature ages in us all, dementia is our fate, sealed by the realit!ies of the circulation; it is not a disease with a cure pending. Empirically, cognitive impairment before our 7th decade is uncommon and considered early, while a diagnosis in our 11th decade is late but common in that cohort (>40%). Projections from earlier ages suggest that the prevalence of dementia in people surviving into their 12th decade exceeds 80%. We address the question why so few of many interventions known to delay dementia are recognized as therapy; and we try to resolve this few-and-many paradox, identifying opportunities for better treatment, especially pre-diagnosis. The idea of dementia as a fate is resisted, we argue, because it negates the hope of a cure. But the price of that hope is lost opportunity. An approach more in line with the evidence, and more likely to limit suffering, is to understand the damage that accumulates with age in the cerebral vasculature and therefore in the brain, and which eventually gives rise to cognitive symptoms in late life, too often leading to dementia. We argue that hope should be redirected to delaying that damage and with it the onset of cognitive loss; and, for each individual, it should be redirected to a life-long defense of their brain.
Collapse
Affiliation(s)
- Marcus J Andersson
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Jonathan Stone
- School of Medical Sciences and Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
22
|
Fišar Z. Linking the Amyloid, Tau, and Mitochondrial Hypotheses of Alzheimer's Disease and Identifying Promising Drug Targets. Biomolecules 2022; 12:1676. [PMID: 36421690 PMCID: PMC9687482 DOI: 10.3390/biom12111676] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/23/2022] [Accepted: 11/09/2022] [Indexed: 08/27/2023] Open
Abstract
Damage or loss of brain cells and impaired neurochemistry, neurogenesis, and synaptic and nonsynaptic plasticity of the brain lead to dementia in neurodegenerative diseases, such as Alzheimer's disease (AD). Injury to synapses and neurons and accumulation of extracellular amyloid plaques and intracellular neurofibrillary tangles are considered the main morphological and neuropathological features of AD. Age, genetic and epigenetic factors, environmental stressors, and lifestyle contribute to the risk of AD onset and progression. These risk factors are associated with structural and functional changes in the brain, leading to cognitive decline. Biomarkers of AD reflect or cause specific changes in brain function, especially changes in pathways associated with neurotransmission, neuroinflammation, bioenergetics, apoptosis, and oxidative and nitrosative stress. Even in the initial stages, AD is associated with Aβ neurotoxicity, mitochondrial dysfunction, and tau neurotoxicity. The integrative amyloid-tau-mitochondrial hypothesis assumes that the primary cause of AD is the neurotoxicity of Aβ oligomers and tau oligomers, mitochondrial dysfunction, and their mutual synergy. For the development of new efficient AD drugs, targeting the elimination of neurotoxicity, mutual potentiation of effects, and unwanted protein interactions of risk factors and biomarkers (mainly Aβ oligomers, tau oligomers, and mitochondrial dysfunction) in the early stage of the disease seems promising.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague, Czech Republic
| |
Collapse
|
23
|
Stecker M. A Perspective: Challenges in Dementia Research. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1368. [PMID: 36295529 PMCID: PMC9609997 DOI: 10.3390/medicina58101368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022]
Abstract
Although dementia is a common and devastating disease that has been studied intensely for more than 100 years, no effective disease modifying treatment has been found. At this impasse, new approaches are important. The purpose of this paper is to provide, in the context of current research, one clinician's perspective regarding important challenges in the field in the form of specific challenges. These challenges not only illustrate the scope of the problems inherent in finding treatments for dementia, but can also be specific targets to foster discussion, criticism and new research. One common theme is the need to transform research activities from small projects in individual laboratories/clinics to larger multinational projects, in which each clinician and researcher works as an integral part. This transformation will require collaboration between researchers, large corporations, regulatory/governmental authorities and the general population, as well as significant financial investments. However, the costs of transforming the approach are small in comparison with the cost of dementia.
Collapse
Affiliation(s)
- Mark Stecker
- Fresno Institute of Neuroscience, Fresno, CA 93720, USA
| |
Collapse
|