Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
Brain ; 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39259179

ABSTRACT

Positive effects of new anti-amyloid-ß (Aß) monoclonal antibodies in Alzheimer's disease (AD) have been attributed to brain amyloid reduction. However, most anti-Aß antibodies also increase the CSF levels of the 42-amino acid isoform (Aß42). We evaluated the associations of changes in CSF Aß42 and brain Aß-PET with cognitive and clinical end points in randomized trials of anti-Aß drugs that lowered (ß- and γ-secretase inhibitors) or increased CSF Aß42 levels (anti-Aß monoclonal antibodies) to test the hypothesis that post-treatment increases in CSF Aß42 levels are independently associated with cognitive and clinical outcomes. From long-term (≥12 months) randomized placebo-controlled clinical trials of anti-Aß drugs published until November 2023, we calculated the post-treatment versus baseline difference in ADAS-Cog (cognitive subscale of the Alzheimer's Disease Assessment Scale) and CDR-SB (Clinical Dementia Rate-Sum of Boxes) and z-standardized changes in CSF Aß42 and Aß-PET Centiloids (CL). We estimated the effect size [regression coefficients (RCs) and confidence intervals (CIs)] and the heterogeneity (I2) of the associations between AD biomarkers and cognitive and clinical end points using random-effects meta-regression models. We included 25 966 subjects with AD from 24 trials. In random-effects analysis, increases in CSF Aß42 were associated with slower decline in ADAS-Cog (RC: -0.55; 95% CI: -0.89, -0.21, P = 0.003, I2 = 61.4%) and CDR-SB (RC: -0.16; 95% CI: -0.26, -0.06, P = 0.002, I2 = 34.5%). Similarly, decreases in Aß-PET were associated with slower decline in ADAS-Cog (RC: 0.69; 95% CI: 0.48, 0.89, P < 0.001, I2 = 0%) and CDR-SB (RC: 0.26; 95% CI: 0.18, 0.33, P < 0.001, I2 = 0%). Sensitivity analyses yielded similar results. Higher CSF Aß42 levels after exposure to anti-Aß drugs are independently associated with slowing cognitive impairment and clinical decline. Increases in Aß42 may represent a mechanism of potential benefit of anti-Aß monoclonal antibodies in AD.

3.
J Neuroinflammation ; 21(1): 187, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080712

ABSTRACT

BACKGROUND: Recent trials of anti-amyloid-ß (Aß) monoclonal antibodies, including lecanemab and donanemab, in early Alzheimer disease (AD) showed that these drugs have limited clinical benefits and their use comes with a significant risk of serious adverse events. Thus, it seems crucial to explore complementary therapeutic approaches. Genome-wide association studies identified robust associations between AD and several AD risk genes related to immune response, including but not restricted to CD33 and TREM2. Here, we critically reviewed the current knowledge on candidate neuroinflammatory biomarkers and their role in characterizing the pathophysiology of AD. MAIN BODY: Neuroinflammation is recognized to be a crucial and contributing component of AD pathogenesis. The fact that neuroinflammation is most likely present from earliest pre-stages of AD and co-occurs with the deposition of Aß reinforces the need to precisely define the sequence and nature of neuroinflammatory events. Numerous clinical trials involving anti-inflammatory drugs previously yielded unfavorable outcomes in early and mild-to-moderate AD. Although the reasons behind these failures remain unclear, these may include the time and the target selected for intervention. Indeed, in our review, we observed a stage-dependent neuroinflammatory process in the AD brain. While the initial activation of glial cells counteracts early brain Aß deposition, the downregulation in the functional state of microglia occurs at more advanced disease stages. To address this issue, personalized neuroinflammatory modulation therapy is required. The emergence of reliable blood-based neuroinflammatory biomarkers, particularly glial fibrillary acidic protein, a marker of reactive astrocytes, may facilitate the classification of AD patients based on the ATI(N) biomarker framework. This expands upon the traditional classification of Aß ("A"), tau ("T"), and neurodegeneration ("N"), by incorporating a novel inflammatory component ("I"). CONCLUSIONS: The present review outlines the current knowledge on potential neuroinflammatory biomarkers and, importantly, emphasizes the role of longitudinal analyses, which are needed to accurately monitor the dynamics of cerebral inflammation. Such a precise information on time and place will be required before anti-inflammatory therapeutic interventions can be considered for clinical evaluation. We propose that an effective anti-neuroinflammatory therapy should specifically target microglia and astrocytes, while considering the individual ATI(N) status of patients.


Subject(s)
Alzheimer Disease , Biomarkers , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Biomarkers/metabolism , Animals , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Precision Medicine/methods
4.
J Alzheimers Dis ; 99(3): 877-881, 2024.
Article in English | MEDLINE | ID: mdl-38701151

ABSTRACT

Three recent anti-amyloid-ß 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-ß42 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.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Peptide Fragments , Humans , Amyloid beta-Peptides/cerebrospinal fluid , Alzheimer Disease/metabolism , Peptide Fragments/cerebrospinal fluid , Risk Assessment , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/therapeutic use
5.
Brain Behav Immun ; 120: 248-255, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38795783

ABSTRACT

Major depressive disorder (MDD) is associated with Alzheimer's disease (AD) but the precise mechanisms underlying this relationship are not understood. While it is well established that cerebrospinal fluid (CSF) soluble levels of triggering receptor expressed on myeloid cells 2 (sTREM2) increase during early stages of AD, how sTREM2 levels behave in subjects with MDD is not known. In a longitudinal study, we measured CSF sTREM2 levels in 27 elderly cognitively intact individuals with late-life major depression (LLMD) and in 19 healthy controls. We tested the hypothesis that, similarly to what happens in early stages of AD, CSF sTREM2 would be elevated in MDD. In addition, we compared the associations of CSF sTREM2, pro- and anti- inflammatory, and AD biomarkers in LLMD and control subjects. Surprisingly, we found that mean CSF sTREM2 levels were significantly reduced in LLMD compared to controls. This reduction was no longer significant at the 3-year follow-up visit when depression severity improved. In addition, we found that CSF sTREM2 was associated with AD biomarkers and proinflammatory cytokines in controls but not in LLMD. These findings suggest that impaired microglia phagocytic response to AD pathology may be a novel link between MDD and AD.


Subject(s)
Alzheimer Disease , Biomarkers , Depressive Disorder, Major , Membrane Glycoproteins , Microglia , Receptors, Immunologic , Humans , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/cerebrospinal fluid , Male , Microglia/metabolism , Female , Aged , Receptors, Immunologic/metabolism , Membrane Glycoproteins/cerebrospinal fluid , Membrane Glycoproteins/metabolism , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/metabolism , Biomarkers/cerebrospinal fluid , Longitudinal Studies , Phagocytosis/physiology , Cytokines/metabolism , Cytokines/cerebrospinal fluid , Middle Aged , Aged, 80 and over , Inflammation/metabolism , Inflammation/cerebrospinal fluid
7.
Ageing Res Rev ; 96: 102290, 2024 04.
Article in English | MEDLINE | ID: mdl-38580173

ABSTRACT

Biomarkers that predict the clinical onset of Alzheimer's disease (AD) enable the identification of individuals in the early, preclinical stages of the disease. Detecting AD at this point may allow for more effective therapeutic interventions and optimized enrollment for clinical trials of novel drugs. The current biological diagnosis of AD is based on the AT(N) classification system with the measurement of brain deposition of amyloid-ß (Aß) ("A"), tau pathology ("T"), and neurodegeneration ("N"). Diagnostic cut-offs for Aß1-42, the Aß1-42/Aß1-40 ratio, tau and hyperphosphorylated-tau concentrations in cerebrospinal fluid have been defined and may support AD clinical diagnosis. Blood-based biomarkers of the AT(N) categories have been described in the AD continuum. Cross-sectional and longitudinal studies have shown that the combination of blood biomarkers tracking neuroaxonal injury (neurofilament light chain) and neuroinflammatory pathways (glial fibrillary acidic protein) enhance sensitivity and specificity of AD clinical diagnosis and improve the prediction of AD onset. However, no international accepted cut-offs have been identified for these blood biomarkers. A kit for blood Aß1-42/Aß1-40 is commercially available in the U.S.; however, it does not provide a diagnosis, but simply estimates the risk of developing AD. Although blood-based AD biomarkers have a great potential in the diagnostic work-up of AD, they are not ready for the routine clinical use.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/metabolism , tau Proteins , Cross-Sectional Studies , Amyloid beta-Peptides , Biomarkers/cerebrospinal fluid
8.
Mol Psychiatry ; 29(3): 847-857, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38228892

ABSTRACT

Alzheimer's disease (AD) is currently constrained by limited clinical treatment options. The initial pathophysiological event, which can be traced back to decades before the clinical symptoms become apparent, involves the excessive accumulation of amyloid-beta (Aß), a peptide comprised of 40-42 amino acids, in extraneuronal plaques within the brain. Biochemical and histological studies have shown that overaccumulation of Aß instigates an aberrant escalation in the phosphorylation and secretion of tau, a microtubule-binding axonal protein. The accumulation of hyperphosphorylated tau into intraneuronal neurofibrillary tangles is in turn correlated with microglial dysfunction and reactive astrocytosis, culminating in synaptic dysfunction and neurodegeneration. As neurodegeneration progresses, it gives rise to mild clinical symptoms of AD, which may eventually evolve into overt dementia. Synaptic loss in AD may develop even before tau alteration and in response to possible elevations in soluble oligomeric forms of Aß associated with early AD. These findings largely rely on post-mortem autopsy examinations, which typically involve a limited number of patients. Over the past decade, a range of fluid biomarkers such as neurogranin, α-synuclein, visinin-like protein 1 (VILIP-1), neuronal pentraxin 2, and ß-synuclein, along with positron emission tomography (PET) markers like synaptic vesicle glycoprotein 2A, have been developed. These advancements have facilitated the exploration of how synaptic markers in AD patients correlate with cognitive impairment. However, fluid biomarkers indicating synaptic loss have only been validated in cerebrospinal fluid (CSF), not in plasma, with the exception of VILIP-1. The most promising PET radiotracer, [11C]UCB-J, currently faces significant challenges hindering its widespread clinical use, primarily due to the necessity of a cyclotron. As such, additional research geared toward the exploration of synaptic pathology biomarkers is crucial. This will not only enable their extensive clinical application, but also refine the optimization process of AD pharmacological trials.


Subject(s)
Alzheimer Disease , Biomarkers , Positron-Emission Tomography , Humans , alpha-Synuclein/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Biomarkers/metabolism , Brain/metabolism , Brain/pathology , Brain/diagnostic imaging , C-Reactive Protein , Nerve Tissue Proteins , Neurocalcin/metabolism , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurogranin/metabolism , Positron-Emission Tomography/methods , Synapses/metabolism , Synapses/pathology , tau Proteins/metabolism
9.
Ageing Res Rev ; 89: 101987, 2023 08.
Article in English | MEDLINE | ID: mdl-37343679

ABSTRACT

Alzheimer's disease (AD) is determined by various pathophysiological mechanisms starting 10-25 years before the onset of clinical symptoms. As multiple functionally interconnected molecular/cellular pathways appear disrupted in AD, the exploitation of high-throughput unbiased omics sciences is critical to elucidating the precise pathogenesis of AD. Among different omics, metabolomics is a fast-growing discipline allowing for the simultaneous detection and quantification of hundreds/thousands of perturbed metabolites in tissues or biofluids, reproducing the fluctuations of multiple networks affected by a disease. Here, we seek to critically depict the main metabolomics methodologies with the aim of identifying new potential AD biomarkers and further elucidating AD pathophysiological mechanisms. From a systems biology perspective, as metabolic alterations can occur before the development of clinical signs, metabolomics - coupled with existing accessible biomarkers used for AD screening and diagnosis - can support early disease diagnosis and help develop individualized treatment plans. Presently, the majority of metabolomic analyses emphasized that lipid metabolism is the most consistently altered pathway in AD pathogenesis. The possibility that metabolomics may reveal crucial steps in AD pathogenesis is undermined by the difficulty in discriminating between the causal or epiphenomenal or compensatory nature of metabolic findings.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/metabolism , Metabolomics/methods , Metabolome , Biomarkers/metabolism
10.
N Engl J Med ; 388(17): 1630-1631, 2023 Apr 27.
Article in English | MEDLINE | ID: mdl-37099353
11.
Alzheimers Dement ; 19(10): 4729-4734, 2023 10.
Article in English | MEDLINE | ID: mdl-37079778

ABSTRACT

Evaluating potential therapies for Alzheimer's disease (AD) depends on use of biomarkers for appropriate subject selection and monitoring disease progression. Biomarkers that predict onset of clinical symptoms are particularly important for AD because they enable intervention before irreversible neurodegeneration occurs. The amyloid-ß-tau-neurodegeneration (ATN) classification system is currently used as a biological staging model for AD and is based on three classes of biomarkers evaluating amyloid-ß (Aß), tau pathology and neurodegeneration or neuronal injury. Promising blood-based biomarkers for each of these categories have been identified (Aß42/Aß40 ratio, phosphorylated tau, neurofilament light chain), and this matrix is now being expanded toward an ATN(I) system, where "I" represents a neuroinflammatory biomarker. The plasma ATN(I) system, together with APOE genotyping, offers a basis for individualized evaluation and a move away from the classic "one size fits all" approach toward a biomarker-driven individualisation of therapy for patients with AD.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/diagnosis , Amyloid beta-Peptides , Biomarkers , Disease Progression , Models, Biological , tau Proteins
12.
Neural Regen Res ; 18(8): 1679-1683, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36751779

ABSTRACT

We reviewed recent major clinical trials with investigational drugs for the treatment of subjects with neurodegenerative diseases caused by inheritance of gene mutations or associated with genetic risk factors. Specifically, we discussed randomized clinical trials in subjects with Alzheimer's disease, Huntington's disease and amyotrophic lateral sclerosis bearing pathogenic gene mutations, and glucocerebrosidase-associated Parkinson's disease. Learning potential lessons to improve future therapeutic approaches is the aim of this review. Two long-term, controlled trials on three anti-ß-amyloid monoclonal antibodies (solanezumab, gantenerumab and crenezumab) in subjects carrying Alzheimer's disease-linked mutated genes encoding for amyloid precursor protein or presenilin 1 or presenilin 2 failed to show cognitive or functional benefits. A major trial on tominersen, an antisense oligonucleotide designed to reduce the production of the huntingtin protein in subjects with Huntington's disease, was prematurely interrupted because the drug failed to show higher efficacy than placebo and, at highest doses, led to worsened outcomes. A 28-week trial of tofersen, an antisense oligonucleotide for superoxide dismutase 1 in patients with amyotrophic lateral sclerosis with superoxide dismutase 1 gene mutations failed to show significant beneficial effects but the 1-year open label extension of this study indicated better clinical and functional outcomes in the group with early tofersen therapy. A trial of venglustat, a potent and brain-penetrant glucosylceramide synthase inhibitor, in Parkinson's disease subjects with heterozygous glucocerebrosidase gene mutations revealed worsened clinical and cognitive performance of patients on the enzyme inhibitor compared to placebo. We concluded that clinical trials in neurodegenerative diseases with a genetic basis should test monoclonal antibodies, antisense oligonucleotides or gene editing directed against the mutated enzyme or the mutated substrate without dramatically affecting physiological wild-type variants.

14.
Pharmacol Res ; 187: 106631, 2023 01.
Article in English | MEDLINE | ID: mdl-36586644

ABSTRACT

According to the ß-amyloid (Aß) hypothesis of Alzheimer's disease (AD), brain Aß accumulation is the primary cascade event leading to cognitive deficit and dementia. Numerous anti-Aß drugs either inhibiting production or aggregation of Aß or stimulating its clearance have failed to show clinical benefit in large scale AD trials, with ß- and γ-secretase inhibitors consistently worsening cognitive and clinical decline. In June 2021, the FDA approved aducanumab, an anti-Aß monoclonal antibody for early AD based on its ability to reduce brain amyloid plaques, while two other amyloid-clearing antibodies (lecanemab and donanemab) have recently produced encouraging cognitive and clinical results. We reviewed AD trials using PubMed, meeting abstracts and ClinicalTrials.gov and evaluated the effects of such drugs on cerebrospinal fluid (CSF) Aß levels, correlating them with cognitive effects. We found that ß-secretase and γ-secretase inhibitors produce detrimental cognitive effects by significantly reducing CSF Aß levels. We speculate that monoclonal antibodies targeting Aß protofibrils, fibrils or plaques may improve cognitive performance in early AD by increasing soluble Aß levels through Aß aggregate disassembly and/or stabilization of existing Aß monomers.These findings suggest that the real culprit in AD may be decreased levels of soluble monomeric Aß due to sequestration into brain Aß aggregates and plaques.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Amyloid beta-Peptides , Cognition
16.
Transl Psychiatry ; 12(1): 301, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35902554

ABSTRACT

Depressed individuals are twice as likely to develop Alzheimer's disease (AD) as compared to controls. Brain amyloid-ß (Aß) deposition is believed to have a major role in AD pathogenesis but studies also suggest associations of Aß dynamics and depression. The aim of this study was to test if plasma Aß levels are longitudinally associated to late-life depression. We measured plasma levels of amyloid-ß1-40 (Aß40) and amyloid-ß1-42 (Aß42) peptides longitudinally for three consecutive years in 48 cognitively intact elderly subjects with late-life major depressive disorder (LLMD) and 45 age-matched cognitively healthy controls. We found that the Aß42/Aß40 plasma ratio was significantly and steadily lower in depressed subjects compared to controls (p < 0.001). At screening, Aß42/Aß40 plasma did not correlate with depression severity (as measured with Hamilton Depression Scale) or cognitive performance (as measured with Mini-Mental State Examination) but was associated to depression severity at 3 years after adjustment for age, education, cognitive performance, and antidepressants use. This study showed that reduced plasma Aß42/Aß40 ratio is consistently associated with LLMD diagnosis and that increased severity of depression at baseline predicted low Aß42/Aß40 ratio at 3 years. Future studies are needed to confirm these findings and examine if the consistently lower plasma Aß42/Aß40 ratio in LLMD reflects increased brain amyloid deposition, as observed in AD subjects, and an increased risk for progressive cognitive decline and AD.


Subject(s)
Alzheimer Disease , Depressive Disorder, Major , Aged , Amyloid beta-Peptides , Biomarkers , Depression/complications , Depressive Disorder, Major/complications , Humans , Longitudinal Studies , Peptide Fragments
17.
Alzheimers Dement ; 18(5): 1008-1037, 2022 05.
Article in English | MEDLINE | ID: mdl-34533272

ABSTRACT

INTRODUCTION: Primary tauopathies are neurological disorders in which tau protein deposition is the predominant pathological feature. Alzheimer's disease is a secondary tauopathy with tau forming hyperphosphorylated insoluble aggregates. Tau pathology can propagate from region to region in the brain, while alterations in tau processing may impair tau physiological functions. METHODS: We reviewed literature on tau biology and anti-tau drugs using PubMed, meeting abstracts, and ClnicalTrials.gov. RESULTS: The past 15 years have seen >30 drugs interfering with tau aggregation, processing, and accumulation reaching the clinic. Initial results with tau aggregation inhibitors and anti-tau monoclonal antibodies have not shown clinical efficacy. DISCUSSION: The reasons for these clinical failures are unclear but could be linked to the clearing of physiological forms of tau by non-specific drugs. Research is now concentrating efforts on developing reliable translational animal models and selective compounds targeting specific tau epitopes, neurotoxic tau aggregates, and post-translational tau modifications.


Subject(s)
Alzheimer Disease , Tauopathies , Alzheimer Disease/pathology , Animals , Brain/pathology , Humans , Tauopathies/pathology , tau Proteins/metabolism
18.
Expert Opin Investig Drugs ; 30(12): 1175-1182, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34890262

ABSTRACT

INTRODUCTION: In the last 15 years, huge efforts against Alzheimer's disease (AD) with drugs targeting ß-amyloid (Aß) and tau have produced poor clinical results. Aducanumab, a recently FDA-approved anti-Aß monoclonal antibody has been greeted with distrust by most experts, hospitals and insurance companies for its level of efficacy and poor tolerability. AREA COVERED: We reviewed literature on Alzheimer trials using PubMed, meeting abstracts and ClnicalTrials.gov and discuss what we can learn from past failures of investigational drugs for Alzheimer's disease, especially anti-Aß and anti-tau drugs. EXPERT OPINION: It is our opinion that previous failures of anti-AD drugs suggest that soluble Aß and tau are not appropriate drug targets. In addition, pivotal clinical trials of future clinical candidates should avoid major protocol amendments and futility analyses. Study protocols should adopt better measures to protect study blinding and minimize the potential introduction of major biases in the evaluation of clinical results. Finally, alternative biological targets should be pursued as well as more multimodal approaches to addressing neurodegeneration in AD.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Amyloid beta-Peptides , Drugs, Investigational , Humans , tau Proteins
19.
J Affect Disord ; 286: 275-281, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33756305

ABSTRACT

BACKGROUND: Decreased cholinergic tone associated with increased proinflammatory cytokines has been observed in several human diseases associated with low-grade inflammation. We examined if this attenuated cholinergic anti-inflammatory pathway (CAP) mechanism contributed to increased neuroinflammation observed in depression. METHODS: We measured cerebrospinal fluid (CSF) cholinergic markers (AChE and BChE activities) in 28 individuals with longstanding late-life major depression (LLMD) and 19 controls and their relationship to central and peripheral levels of pro-inflammatory cytokines (IL-6 and IL-8). Additionally, we examined if these cholinergic indices were related to CSF markers of microglial activation and neuroinflammation (sTREM2 and complement C3). RESULTS: Compared with controls, LLMD patients had a significant reduction in CSF BChE levels. Lower CSF BChE and AChE activities were associated with lower CSF markers of microglial and neuroinflammation (sTREM2 and C3). In addition, in LLMD patients we found an inverse relationship between peripheral marker of inflammation (plasma IL-6) and CSF BChE and AChE levels. CONCLUSIONS: Our results suggest an upregulation of the CAP mechanism in LLMD with an elevation in peripheral markers of inflammation and concomitant reduction in markers of glial activation associated with a higher cholinergic tone. Future studies should confirm these findings in a larger sample including individuals with acute and more severe depressive episodes and across all ages.


Subject(s)
Depression , Neuroimmunomodulation , Acetylcholinesterase/metabolism , Cholinesterases , Humans , Up-Regulation
20.
Expert Opin Drug Discov ; 16(7): 727-735, 2021 07.
Article in English | MEDLINE | ID: mdl-33653187

ABSTRACT

Introduction: As the global burden of Alzheimer's disease (AD) grows, an effective disease-modifying therapy remains a distant prospect following the repeated failure of multiple therapeutics targeting ß-amyloid and (it seems) tau over many years of costly effort. The repeated failure of single-target therapies to meaningfully modify disease progression raises major questions about the validity of many aspects of drug development in this area, especially target selection.Area covered: The authors explore the critical questions raised by a review of the collective experience to date, relating to why findings with non-clinical models and clinical biomarkers so frequently fail to translate to positive outcomes in clinical trials, which alternatives should be considered, and how we can design and conduct clinical trials that can successfully identify and quantify meaningful benefits in the future.Expert opinion: It is our opinion that we must recognize and accept the need to consider less specific, more multimodal approaches to addressing neurodegeneration in AD if we are to make progress - and we must avoid repeating the well intentioned, but ultimately erroneous, assumptions of the past.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Amyloid beta-Peptides , Disease Progression , Drug Development , Drug Discovery , Humans
SELECTION OF CITATIONS
SEARCH DETAIL