• Open Access
    Review

    Current therapeutics for Alzheimer’s disease and clinical trials

    Danqing Xiao 1*
    Chen Zhang 2,3*

    Explor Neurosci. 2024;3:255–271 DOI: https://doi.org/10.37349/en.2024.00048

    Received: March 15, 2024 Accepted: May 22, 2024 Published: June 27, 2024

    Academic Editor: Ryszard Pluta, Medical University of Lublin, Poland

    This article belongs to the special issue Alzheimer’s Disease

    Abstract

    Alzheimer’s disease (AD) is a major type of dementia and neurodegenerative disease, characterized by memory loss and cognitive decline. Over decades, significant efforts have been dedicated to finding its cause, pathogenic mechanisms, biomarkers for early detection, and clinical trials for its treatment. Earlier approved drugs mainly ameliorated the symptoms of AD, until recent years when two drugs targeting amyloid-beta (Aβ) protein were approved to slow down the progression of the disease. This review article encompasses the history of drug development in treating AD and clinical trials that failed and succeeded. Clinicaltrials.org website was systematically searched and screened for randomized controlled trials with results posted in the past 10 years. Among the 3,388 AD clinical trials, 211 interventional studies registered under AD have met eligibility. This review includes the interventional targets for drug discovery such as Aβ, tau, neurotransmitter receptors, neuroinflammation, multi-target studies, repurposing pharmacological agents, non-pharmacological interventions, and clinical therapy development for the neuropsychiatric symptoms of dementia. Current clinical trials are ongoing and no results are available as of yet. With the vast choices of drug targets that have been investigated, this review aims to present some insights into future AD drug design and trials and contribute to our ongoing efforts to find the cure.

    Keywords

    Clinical trials, Alzheimer’s disease, amyloid-beta, tau protein, immunotherapies, multi-target drugs

    Introduction

    Alzheimer’s disease (AD) has affected around 50 million people worldwide and is projected to reach 152 million by 2060 (for a review, see [1, 2]). AD and other dementias ranked among the top 10 leading causes of death globally [3]. Currently, there are about 24 million Alzheimer’s patients worldwide [2]. It was suggested that gene changes can be a 70% risk factor for AD development [4, 5]. Three genes, also the main (high) risk factors for AD, are associated with early-onset AD. They are APP on 21q, PSEN1 on 14q and PSEN2 on 1q [6]. The apolipoprotein E (APOE) gene is the strongest genetic (medium) risk factor for late-onset AD, located on chromosome 19q [7]. Among the APOE gene variants, APOE4 is considered the strongest genetic risk factor for AD, and APOE2 is neuroprotective [8].

    FDA-approved immunotherapy drugs targeting amyloid-beta (Aβ) work best to slow the cognitive declining at the early stages of AD for some people, unlike earlier approved cholinergic drugs that provide symptomatic relief without altering the disease progression. The majority of clinical trials are targeting intervention with AD therapeutic agents at early/mild to moderate stages of AD, and its early detection. This review aims to provide an overview of the current treatments for relieving some AD symptoms by targeting cholinergic and N-methyl-D-aspartic acid (NMDA) receptors, and clinical trials in halting the progression of AD, targeting removing the toxic proteins typical of AD, Aβ proteins (extracellular plaques) and various forms of tau proteins (intracellular tangles) or other targets and multi-target drugs.

    Approved treatments for Alzheimer’s disease

    Acetylcholinesterase inhibitors

    The mainstay of current AD management targeting acetylcholinesterase is to make acetylcholine available for synaptic transmission [2, 911]. Five drugs (donepezil, rivastigmine, galantamine, memantine, and tacrine) are approved on the market with donepezil approved the earliest, in 1996. These drugs are used for all stages of AD, but inhibition of cholinesterase enzymes works best at the early stage of AD [9, 12].

    Among them, a rivastigmine patch or oral capsule is the only acetylcholinesterase inhibitor (ACEI) that provides inhibition for both acetylcholinesterase and butyrylcholinesterase and shows efficacy and safety in helping to relieve the cognitive symptoms of AD (e.g., [13]).

    NMDA receptor antagonist

    Current therapeutics to ameliorate and control the cognitive and behavioral symptoms by targeting the NMDA receptor prevent Ca2+ induced excitotoxicity [2, 911], and memantine was approved for this purpose in 2003. Memantine is more effective in providing cognitive and behavioral benefits and approved for patients with moderate to severe AD as monotherapy or combination therapy with donepezil [9, 12]. Finally, memantine is both a non-competitive NMDA receptor antagonist and a dopamine agonist for moderate to severe AD [11].

    Anti-amyloid monoclonal antibodies

    The FDA has approved two drugs for mild cognitive impairment (MCI) to early, mild stages of AD after years of failed clinical trials. They are aducanemab, and lecanemab, among which aducanemab received accelerated approval [14, 15].

    Clinical trials for Alzheimer’s disease

    As of May 2024, there are a total of 3,388 Alzheimer’s disease clinical trials including all studies (clinicaltrials.gov). We chose to focus on the trials starting from 01/01/2015 and encompassing the past 10 years, including 2,030 studies. After excluding observational studies (with patient registries), and keeping all the interventional clinical trials, there are 1,596 studies registered under AD. After keeping the trials with results (excluding the ones without results), 211 studies remain with (early) phase 1 (n = 32), phase 2 (n = 78) trials on the safety and tolerability test, and phase 3 (n = 37) and phase 4 (n = 4) trials on slowing the cognitive decline. These trials were or are in the process of being carried out globally. This review includes the interventional targets for drug discovery such as neurotransmitter (receptors), Aβ, tau, neuroinflammation, growth factors, and hormones, including multi-target studies (Table 1, 2, 3); repurposing pharmacological agents already in use; non-pharmacological interventions such as environmental enrichment, combination therapy, and neurostimulation (devices); and clinical therapy development for the neuropsychiatric symptoms of dementia based on Common Alzheimer’s and Related Dementias Research Ontology (CADRO). Anti-inflammatory therapy is intended to reduce the harmful inflammatory response of microglia activation in the AD brain [1618]. Moreover, there are other important approaches, for example, the exploration and efforts targeting oxidative stress linked to mitochondria dysfunction to screen neuroprotective antioxidant compounds in cells and mouse models of AD [16, 17, 19].

    Compounds target for cognitive improvement in AD and their combination treatments

    NameTarget and rationaleStatus on clinical trials (start date–end date)AD stageResultsClinical trials identifier (NCT number)
    CT1812Antagonists for the sigma 2 and progesterone receptor membrane component 1 (PGRMC1) receptorsPhase 1–2 (2018–2020)Mild to moderateNo alteration of synaptic density, insignificant changes in cognition memory task03493282
    Rasagiline (Azilect)MAOB inhibitor, reducing reactive astrocytesPhase 2 (2015–2019)Mild to moderateGlucose metabolism declining less showed benefit in quality of life only02359552
    LadostigilAchE inhibitor and MAO inhibitor, promoting cholinergic and serotonergic/dopaminergic neurotransmissionPhase1/2 (2019)MildFailed to meet the primary endpoint01429623
    IdalopirdineAchE inhibitor and 5-HT6 receptor, stimulating 5-HT and cholinergic receptor (as an adjunct therapy for AchE inhibitor)Phase 1–3 (2014–2017)Mild to moderateWeak efficacy in phase 3, discontinued (2017)02079246
    RivastigmineInhibitors of AchE and butyrylcholiesterasePhase 4 (2016–2018)Mild to moderateShowing some symptomatic improvement [13]02703636
    Intepirdine (RVT-101)The antagonist of the 5-HT6 receptor, CNS-specificPhase 2 (2016–2017)
    Phase 3 (2016–2018)
    Mild to moderateDid not meet primary efficacy endpoints02586909
    02910102
    BI 409306Inhibitor of phosphodiesterase (PDE) 9A, increasing cGMPPhase 2 (2015–2017)ProdromalNo difference between drug and placebo groups on measures of efficacy02240693
    02337907
    Neflamapimod (VX-745)Inhibitor of p38 MAPK (mitogen-activated protein kinase) alphaPhase 2 (2015–2016)
    Phase 2 (2017–2019)
    MildFailed to meet the primary endpoint of improving episodic memory02423122
    03402659
    BI 425809Inhibitor of glycine transporter, keeping a higher level of glycine at the synapse promoting NMDA R activationPhase 2 (2016–2019)Mild to moderateNo improvement in the primary or secondary measures compared with the placebo [20]02788513
    CandesartanBlocker of Angiotensin IIPhase 2 (2016–2020)MCISuperior in primary and secondary outcomes of executive function by TMT(B) and Hopkins Verbal Learning Test-Revised delayed recall [21]02646982
    Donepezil plus solifenacin (CPC-201)Inhibitor of AchE and muscarinic receptor M3 treating overactive bladder, respectivelyPhase 2 (2015–2017)ModerateAllowing the safe use of higher AchE inhibitors doses that may augment cognitive and global benefits [22]02549196
    Donepezil + memantine + masupirdine (SUVN-502)Antagonists of the AChE, NMDA R, and 5-HT6 receptorPhase 1–2 (2018)ModerateNo additional benefit with the combination treatments02580305
    Memantine + bryostatinNMDA R antagonist, macrolide lactones which increase the α-secretase activity and reduce Aβ accumulationPhase 2 (2015–2017)Moderate to severeNo additional benefit with the combination treatments02431468
    Display full size

    AD: Alzheimer’s disease; 5-HT6: 5-hydroxytryptamine 6; MAOB: monoamine oxidase-B; MCI: mild cognitive impairment; TMT(B): Trail-Making Test-B; Aβ: amyloid-beta; NMDA R: N-methyl-D-aspartic acid receptor

    Status and rationale of selected compounds targeting various amyloid proteins tested in AD clinical trials

    Invested agent nameTarget and rationaleStatus on clinical trials (start date–end date)AD stageResultsClinical trial identifier (NCT number)
    Posiphen tartrate (Buntanetap)APP synthesis reductionPhase 1–2 (2021)Mild to moderateTerminated due to business decision04524351
    Bryostatinα-Secretase activity enhancer, reducing AβproductionPhase 2 (2019)Moderate to severeIneffective03560245
    LY3202626β-Secretase activity reduction [targeting BACE1 (β site APP cleaving enzyme 1)]Phase 2 (2018)MildTerminated due to low likelihood of identifying a statistically significant treatment effect02791191
    Lanabecestat (LY3314814)β-Secretase activity reduction (targeting BACE1)Phase 2/3 (2018)MildLack of efficacy or terminated due to unlikely to meet the primary endpoint and brain volume reduction02245737
    02783573
    Verubecestat
    (MK-8931)
    β-Secretase activity reduction (targeting BACE1)Phase 2–3 (2017)Mild to moderateTerminated due to injuries and lack of efficacy01739348
    Umibecestat
    (CNP520)
    β-Secretase activity reduction (targeting BACE1)Phase 2/3 (2019 and 2021)Mild, or at risk of AD with one allele of APOE4 and elevated brain amyloidWorsening cognition, and terminated due to safety reasons03131453
    Elenbecestat (E2609)β-Secretase activity reduction (targeting BACE1)Phase 3 (2020)Mild/EarlyEnded Due to an unfavorable risk-benefit ratio including lack of efficacy, and the adverse event being worse than the placebo with brain volume loss02956486
    Semagacestatγ-Secretase inhibitor, preventing Aβ production (not selective)Two phase 3 trials (2017–2020)Mild to moderateSignificant deterioration of cognitive function, side effects (GI and skin cancer)02423122
    03402659
    ELND005Inhibiting Aβ formationPhase 2 (2012–2015)ModerateAdverse effect (infection)01735630
    Exendin4A glucagon-like peptide, binding to soluble Aβ fragments, lowering Aβ burdenPhase 2 (2016)Prodromal/MildAdverse effect (infection), terminated due to lack of support01255163
    (Azeliragon) TPP488RAGE (cell surface receptor linked to an increase in Aβ plaque formation) antagonist, increase Aβ clearance and targeting neuroinflammationPhase 2 (2019–2021)
    Two phase 3 trials (2015–2018, 2016–2018)
    MildPrimary endpoints not met03980730
    02080364
    02916056
    PioglitazoneAgonizing peroxisome proliferator-activated receptor (PPAR) to increase Aβ phagocytosisPhase 3
    (2015–2018)
    MCITerminated due to lack of efficacy02284906
    Crenezumab (MABT5102A, RG7412)Human monoclonal antibody (hmAB) targeting the clearance of Aβ oligomers and fibrillar speciesPhase 3 (2016, 2017, 2018–2019)Prodromal to mildDiscontinued by the sponsor due to lack of efficacy [23]02670083
    03114657
    03491150
    SolanezumabhmAB targeting soluble Aβ peptidePhase 3 (2013–2017)Mild to moderateLack of efficacy01900665
    GantenezumabhmAB binds to insoluable Aβ plaques and removes them via phagocytosisPhase 3
    (2010–2020,
    2014–2021)
    Mild/ProdromalLack of efficacy, its primary endpoints not met01224206
    02051608
    Aducanumab (BII3037)hmAB targeting the clearance of Aβ plaquesPhase 2 (2018–2019)
    Phase 3 (2015–2019, 2015–2019, 2020–2024, 2022–2024)
    Mild/EarlyTwo identical trials (ENGAGE and EMERGE) were terminated due to futility analysis. Substudies for both trials showed a dose-dependent reduction of Aβ and CDR-SB. In the EMERGE trial (NCT 02484547), aducanemab at a high dose significantly reduced dementia rating score CDR-SB. The trial (NCT05310071) would be terminated due to the sponsor’s decision03639987
    02484547
    02477800
    04241068
    05310071
    Donanemab (LY3002813)hmAB targeting the clearance of Aβp3-42 plaquesPhase 1–3
    (2017–2021, 2023, 2020–2024)
    Mild/EarlyReduced Aβ plaques and tau, are under review for standard approval [14]03367403
    04437511
    05108922
    04640077
    Lecanemab (BAN2401)hmAB targeting the clearance of soluble Aβ aggregatesPhase 1–3
    (2012–2023 2019–2027)
    MildReduced Aβ plaques and tau, with less decline of cognition and function. Received full FDA approval02094729
    03887455
    UB-311
    (AD vaccine)
    Active immunotherapy with Aβ peptide (N-terminal amino acid 1-14), inhibiting Aβ aggregatesPhase 2 (2015–2018)MildRobust immune response with adverse events such as microhaemorrages (14%), supporting phase 3 trials [24]02551809
    Display full size

    AD: Alzheimer’s disease; Aβ: amyloid-beta; APP: Aβ precursor protein; MCI: mild cognitive impairment; CDR-SB: clinical dementia rating-sum of boxes; GI: gastrointestinal; APOE 4: apolipoprotein E 4

    Status and rationale of selected compounds targeting various tau proteins tested in AD clinical trials

    Invested agent nameTarget and RationaleStatus on clinical trial
    (start date–end date)
    AD stageResultsClinical trial identifier (NCT number)
    ABT-957 (Alicapistat)Inhibition of calcium-dependent cysteine protease, calpain, which is linked to hyperphosphorylation of tau and its aggregationPhase 1, (2014–2016)Mild to moderateNot reaching the effective level in CNS [25]02220738
    Semorinemab, RO7105705, RG6100IgG4 antibodies targeting all 6 isoforms of extracellular tau (eTau)Phase 2, (2021)Mild/ProdromalThe primary endpoint not met didn’t show benefit or modulation of the tau-PET signal03289143
    Phase 2, (2023)Probable AD and moderatePromising results on cognition03838747
    BIIB092,
    Gosuranemab
    hmAB against eTauPhase 1/2, (2018–2021)MildLack of efficacy03352557
    Zagotenemab, LY3303560hmAB against a conformational epitope of eTauPhase 1/2, (2016–2021)Mild or early symptomaticMissed primary endpoints03518073
    RG7345,
    RO6426496
    Rabbit monoclonal Ab targeting phosphorylated Tau (at serine 422)Phase 1, (2015)Healthy malesTerminated, unfavorable phamacokinetic profile02281786
    BIIB076,
    NI-105
    Human recombinant monoclonal anti-tau Ab targeting mid-domain of TauPhase 1 (2017–2020)Healthy and mildDiscontinued due to business reasons03056729
    AADvac1Active vaccine using a synthetic tau peptidePhase 1
    (2013–2015)
    Phase 2
    (2016–2019)
    Mild to moderateSafe [26]
    and promising, but lack efficacy [27]
    01850238
    02579252
    Display full size

    AD: Alzheimer’s disease; hmAB: human immunoglobulin monoclonal antibody

    Further, combination therapy can combine both pharmacological approaches and/or combine non-pharmacological approaches. Combination therapy that improves symptoms plus disease-modifying therapeutics (DMT) would modify and slow down the progression of AD as add-on treatments in combination with the current drugs. This combination therapy gives flexibility, gearing towards individual differences in regards to the drug target or multiple targets, more than one delivery method or delivery timing (sequential combinations), and the use of multifunctional molecules-single agents (multi-target drugs) that combine more than one activity or more than one target [28]. Combination therapy can also combine pharmacology and non-pharmacological intervention, such as gamma frequency neurostimulation (see the following section: non-pharmacological interventions and [28]).

    Multi-target drugs acting on different targets

    To improve efficacy, single molecule drugs have been designed to incorporate two or more pharmacological structural features of different bioactive drugs acting on different targets. The different structural components can be connected through a tether/linker or merged together [9]. Examples include ANAVAX 2-73 (blarcamesine), lumateperone, idolopiridine, ladostigil, and J-147.

    Blarcamesine, combining agonist activities at the muscarinic M1 receptor, the NMDA receptor, and the sigma-1 receptor, targets protein tau and reduces AD neurodegeneration [29]. Two consecutive multicenter phase 2a trials, randomized and open-label, show it improves cognition, dose-dependently, in mini-mental status examination (MMSE) and AD Cooperative Study-Activities of Daily Living Scale (ADCS-ADL) in subjects with mild-to-moderate AD [30]. The ongoing ATTENTION-AD trial (NCT04314934) is a phase 2b/3 open-label extension study to evaluate the effects of blarcamesine on the safety and efficacy of daily treatment with an enrollment of 450 participants globally who have completed the previous ANAVEX2-73-AD-004 double-blind study (clinicaltrials.gov). Lumateperone and iolopiridine targeted both 5-hydroxytryptamine 6 (5-HT6) and AchE, though both were discontinued due to lack of efficacy. Ladostigil is a hybrid molecule combining the carbamate group of rivastigmine and the N-propargyl group of monoamine oxidase (MAO) inhibitors, though it failed to meet the primary endpoint of delaying cognitive impairment in phase 2. J-147 under phase 1 is a multi-target drug including targets like MAO. MAOB activity is increased in AD at reactive astrocytes near amyloid plaques.

    Clinical trials targeting Aβ protein

    The rationales of these early AD drug trials from 2011 to 2020 are based on the Aβ hypothesis. Trials targeting degradation and production from Aβ precursor protein (APP) include those designed to: (1) decrease APP production, for example, posiphen (buntanetap); (2) increase α-secretase (stimulating non-amyloidogenic pathway), such as bryostatin; (3) inhibition of β-secretase to reduce Aβ production, in amyloidogenic pathway) [9], such as atabecestat and lanabecestat [31]; (4) inhibition of gamma-secretase (to reduce Aβ production, in the amyloidogenic pathway), such as LY3202626; (5) target Aβ solubility and aggregation such as Alzhemed (discontinued) and its pro-drug ALZ-801(for its potential efficacy in patients with the APOE4/4 genotype or its carriers), as soluble Aβ aggregates are considered to be the relevant cytotoxic form of the protein [32]; (6) target Aβ clearance by antagonizing RAGE (cell surface receptor linked to an increase in Aβ plaque formation), which primarily targeting neuroinflammation. These trials resulted in failure due to lack of efficacy and/or side effects of toxicity, for example [31] (Table 2).

    Aβ clearance by immunotherapy was first demonstrated in an ischemic mouse model showing amyloid plaques can disappear with injections of human β-amyloid-(1-42)-peptide for some months [33]. Aiming for elimination of Aβ aggregation has two approaches: passive immunization and active immunization. Passive immunotherapies with monoclonal antibodies against Aβ in clinical trials went through three generations of development [11, 34], with FDA approval of two drugs for MCI to early stage of AD. First-generation antibody against N-terminus aggregated amyloid plaque, bapineuzumab, has no efficacy, due to low potency [35]. Second-generation anti-amyloid antibody against soluble monomeric Aβ (solanezumab), insoluble monomeric Aβ (gantenezumab), and monoclonal antibody against multiple Aβ forms (crenezumab) failed to have efficacy in improving cognitive functions for mild and moderate AD in several clinical trials (Table 2, for example [36]). The third generation of human immunoglobulin monoclonal antibody (hmAB; aducanumab, donanemab, and lecanemab) directed against insoluble, modified, N-terminal truncated form of Aβ, targeting Aβ plaque clearance, seems to benefit MCI or AD with mild dementia patients [34, 37]. Aducanumab and lecanemab are the first agents in two decades to demonstrate efficacy in clinical studies.

    Aducanemab’s benefits are controversial [38, 39]. Four aducanumab phase 3 trials against Aβ fibrils were carried out; two were terminated due to futility analysis, and two trials (ENGAGE and EMERGE) showed benefit by reducing AD biomarkers, Aβ and tau proteins, in early-onset AD in certain patients [9]. Sub-studies for the two trials showed a dose-dependent reduction of Aβ and clinical dementia rating-sum of boxes (CDR-SB) a global measure of both cognition and function. In the EMERGE trial (NCT 02484547), aducanumab at a high dose significantly reduced dementia rating score CDR-SB (clinicaltrials.org, for a review see [14]). In 2024, the marketing of aducanumab was discontinued by the manufacture. Donanemab (under FDA review) can effectively reduce the cerebral spinal fluid (CSF) level of amyloid protein and plasma P-tau and T-tau [40, 41], provides a benefit of slowing down cognitive declines and improving function, and meet the primary endpoint, the integrated AD Rating Scale (IADRS), for those who are at the earliest stage of AD with a low level of tau proteins, and to a lesser extent those who carry APOE4 [42]. Lecanemab, directed against amyloid protofibrils, is approved for MCI/mild AD dementia based on the clarity AD trial (NCT03887455). This is an 18-month, multicenter, double-blind, phase 3 trial with early AD (MCI or mild dementia due to AD) with evidence of amyloid on PET or CSF testing. The administration of this antibody manifested significant (27%) slower progress in cognitive decline in the patients based on the primary end points, the change from baseline at 19 months in the score on the CDR-SB (with higher scores being greater impairment), and secondary endpoints, the change in the 14-item cognitive subscale of the AD assessment scale (ADAS-cog 14; higher scores being greater impairment), the AD composite score (ADCOMS; higher being worse) and the score on the ADCS-ADL for MCI (ADCS-MCL-ADL; lower scores being greater impairment). Overall, lecanemab significantly reduced amyloid burden with modestly less decline in measures of cognition and function in early-stage AD patients [34, 39, 43].

    For cognitively normal individuals who are at high risk carrying AD genes, clinical trials were also started to prevent AD from onset or delay its onset. In one AD trial, crenezumab was tested for its preventative effect. The benefits of crenezumab on cognitively unimpaired autosomal dominant AD participants wait to be understood based on the recently closed phase 2 clinical trial (NCT 01998841) [44, 45].

    Finally, trials on active immunotherapies targeting Aβ are limited. The first active immunization in humans, with the AN 1792 vaccine, was terminated due to its ineffectiveness in removing Aβ deposits from the brain or slowing cognitive decline, and adverse effects [46]. Later vaccine development resulted in improvement of cognition and reduction of Aβ accumulation; for example, active AD vaccine UB-311 has shown promising results for mild AD patients, able to generate a high response rate (100%) of antibody production in a phase 2 trial, with plans for a clinical phase 3 trial (Table 2) [9, 47].

    Clinical trials targeting tau proteins

    Although amyloid remains the most common pharmaceutical target, targeting the tau protein, which is downstream to Aβ oligomers and contributes to intracellular neurofibrillary tangles, has received attention in recent years (Table 3; Clinicaltrials.org) [14, 48]. A tau imaging study showed tau accumulates slowly in large neocortical areas, taking approximately 15–20 years, with the highest rates of tau accumulation at the temporal, parietal, and lateral occipital cortex [49]. Currently, there is more evidence that hyperphosphorylated tau plays an important role in AD pathogenesis, reducing synaptic transmission at the nerve terminals and leading to tangle formation [37].

    So far, three generations of anti-tau non-immunotherapy drugs failed clinical trials. First-generation anti-tau drugs target reversing post-translation hyperphosphorylation modifications that induce tau aggregation or inhibit tau aggregation directly, for example, nicotinamide and curcumin [50]. However, the tau aggregation inhibitor leuco-methylthioninium bis (hydromethanesulphonate), (LMTM, TRX 0237) trial, treating mild and moderate AD and frontotemporal dementia (2018), was terminated and failed at both primary endpoints (Alzforum.org). Second-generation anti-tau, Saracatinib (AZD0530), is a dual kinase inhibitor of Src and Ab1 family kinases and a repurposed drug originally treating cancer. Third-generation anti-tau drug Atuzagnstat (COR 388), for mild to moderate AD (2019–2022), failed due to adverse gastrointestinal (GI) effects and subject withdrawal.

    On the other hand, the anti-tau immunotherapeutic approach is more complicated due to the various isoforms of tau protein, with at least six monoclonal antibodies against tau now in phase 2 and phase 3 clinical trials [11, 51]. Earlier phase 1 clinical trials targeting tau failed due to lack of recruitment; these agents reduce tau build-up via histamine H3 receptor antagonists and calcium-dependent cysteine protease inhibitors for mild to moderate AD. Later anti-tau therapeutic design includes antibody against different forms of tau proteins, mostly targeting the N-terminus such as semorinemab and gonsuranemab, or microtubule-binding regions such as AADvac1 and zagotenemab which are shown to reduce CSF P-tau level [50] (Table 3). Semorinemab and R07105705 are active against all six isoforms of tau, whereas, RG7345 is only active against phosphorylated-tau. Semorinemab was ineffective and did not slow the decline in a primary cognitive outcome for early AD in the phase 2 trial [52].

    Active vaccine AADvac1, a synthetic tau peptide, reduces tau deposit by attacking the conformational epitope of tau for mild to moderate AD and has been well tolerated in clinical trial phase 1–2. A subgroup analysis of younger AD patients (participants 67 years or younger) showed significant beneficial changes in biomarker plasma neurofilament light chain (NfL) and cortical atrophy [51]. A post hoc subgroup analysis of the participants positive for amyloid and tau from phase 2 trial (NCT02579252) found the vaccine slowed AD-related decline on the CDR-SB and ADL scores in an antibody-dependent manner, and brain volume was preserved with treatment compared with control [48]. Phase 3 trial of active vaccine against tau protein is anticipated for treating mild AD [51].

    Moreover, the antibody against tau protein can recognize extracellular tau (in most cases) or intracellular tau, or both. Antibody recognizing extracellular tau is thought to block the spread of tau across synapses [53], though it may not be perfect, for example, BIIB092, CN2-8E12, tilavonemab, and LY3303560 that recognize extracellular tau protein lack efficacy (Table 3). To target production and degradation of the intracellular tau, antisense oligonucleotides and small interfering ribonucleic acids (siRNAs) treatment is used to reduce tau production. In addition, proteolysis-targeting chimeras are used to enhance tau proteasomal degradation in early AD [50]. A current phase 2 clinical trial is investigating whether upstream gene activation, triggering receptor expressed on myeloid cells 2 (TREM-2) dependent microglia activation, can halt tau accumulation inside the cells (NCT04592874). One might assume that an antibody recognizing both extracellular and intracellular tau protein will be the most efficacious. However, this is not the case; for example, RG7345 that can bind to both intracellular and extracellular tau proteins has an unfavorable pharmacokinetic profile (Table 3).

    Repurposing pharmacological agents

    Repurposing pharmacological agents has advantages from a drug development standpoint. Based on FDA regulation, an application that contains reports of safety and effectiveness in at which at least some of the information required for approval comes from studies conducted for a previously approved product receives much shortened review, leading to a speedy approval compared to a new drug. There are numerous repurposed drug trials that had encouraging results. Masitinib targets (inhibiting) tyrosine kinase and modulated neuroinflammation in 2012–2020 phase 2/3 trials for mild to moderate AD [9]. Minocyclin is a repurposed drug on trial for mild AD. Riluzole, a D2 receptor agonist approved for ALS, completed its phase 2 trial for mild AD as well and showed promise in improving glucose metabolism [54]. Rasagiline, an MAOB inhibitor approved for Parkinson’s disease, showed a favorable effect on neuroimaging biomarker tau and some benefit in quality of life in patients with mild to moderate AD [55]. Nabilone, an antiemetic and synthetic cannabinoid completed a phase 3 trial in 2019 and showed its promise in controlling agitation, a challenging and the most prevalent neuropsychiatric symptom, through anti-inflammatory effects in moderate-to-severe AD patients [56, 57].

    Pharmacological combination therapy and ongoing clinical trials

    Considering the complexity of AD pathogenesis, using combination drug therapy on two or more different targets presents the potential for improved efficacy and outcomes, as targeting multiple pathways or mechanisms might have a synergistic effect on therapeutic outcomes. For example, Namzaric, a fixed dose-combination of donepezil and memantine, is used for the treatment of AD patients with moderate to severe AD dementia [9]. However, adding the 5-HT6 receptor antagonist, masupirdine (SUVN-502), as adjuvant to donepezil and memantine, demonstrated no additional palliative benefits in a phase 2 clinical trial (NCT02580305) [58, 59]. It was suggested that memantine interferes with the effect of 5-HT6 antagonist masupirdine.

    Other pharmacological combination therapy includes targeting both neuroinflammation and Aβ clearance as in t-octhylphenol 1 (T-OP1), combining a mast cell stabilizer and an Aβ aggregation inhibitor (cromolyn) with a nonsteroidal anti-inflammatory drug (NSAID) [60], and targeting tau (with antibody or a microtubule stabilizer) and acetylcholinesterase inhibitors (AChEI) therapy, the latter of which worked better when targeting mild to moderate stage AD in clinical trial. However, most of these trials failed to meet each trial’s specific cognitive endpoints, for example, clinical trials of Aβ clearance agent Rosiglitazone as an add-on therapy for patients with AD on donepezil or other AChEIs [37].

    Current research suggests that amyloid and tau have a synergistic interaction and effect in promoting neurodegeneration and cognitive decline [61], and therefore, clinical trials that target both removing Aβ protein and tau burden with lecanemab are ongoing with the results yet to be released (Table 4). Other ongoing clinical trials aim to remove Aβ plaques with hmAB such as aducanumab, donanemab, gantenezumab, and lecanemab, and continuously monitor their safety and efficacy. Lastly, with the hope that tau is a promising candidate, ongoing clinical trials target intracellular tau and different regions of extracellular tau to block the spread of prion-like tau, for example, JNJ63733657 and UCB0107 (Bepranemab) targeting microtubule binding and tau mid-region, respectively (Table 4).

    Status and rationale of ongoing AD clinical trials

    Invested agent nameTarget and rationaleStatus on clinical trial
    (phase, start date–end date in year)
    AD stageClinical trial identifier (NCT number)
    ANAVAX 2-73
    (Blarcamesine)
    Sigma 1 receptor agonist, modulating muscarinic receptors to decrease the APP synthesis, restoring homeostasisPhase 2/3 (2019–2024 July)Early04314934
    ALZ-801
    (a prodrug version of Alzhemed)
    Binding to soluble Aβ fragments, inhibiting Aβ aggregationPhase 2 (2020–2024)
    Two phase 3 trials (2021–2024, 2024–2026)
    MCI-moderate
    Early AD with the APOE4 carriers, or APOE4/4 genotype
    04693520
    04770220
    06304883
    AducanumabHuman anti Aβ monoclonal Ab targeting the clearance of Aβ plaquesPhase 3 (2020–2025)Mild04241068
    DonanemabHuman anti Aβ monoclonal Ab targeting the clearance of Aβp3-42 plaquesPhase 3 (2021–2027)
    Phase 3 (2020–2025, 2023–2025, 2022–2027)
    Preclinical
    Early
    05026866
    04437511
    05738486
    05508789
    Lecanemab
    (BAN2401)
    Human anti Aβ monoclonal Ab targeting the clearance of soluble Aβ aggregatesPhase 2–3 (2012–2027)Preclinical/Early01767311
    03887455
    04468659
    Phase 2–3 (2012–2027)Mild or cognitive normal EOAD05269394
    01760005
    AL002Halting intracellular tau accumulation via TREM-2 dependent microglia activationPhase 2 (2021–2024 September, 2023–2025)Early AD04592874
    05744401
    JNJ-63733657Human IgG1 monoclonal Ab recognizes the microtubule binding region of phosphorylated tau (p217+), interferes with spread of pathogenic tauPhase 2 (2021–2025)Early AD04619420
    UCB0107
    Bepranemab
    Human IgG4 monoclonal Ab recognizing the mid-region of tau (aa. 235-250), near the microtubule-binding domain, interferes with the spread of tau, more potent than N-terminally targeted antibodiesPhase 2 (2021–2025)Mild or prodromal04867616
    Lecanemab with and w/o E2814Anti-Ab and Tau antibody therapy to reduce the burden of bothPhase 2–3 (2021–2027)Cognitively normal or MCI, with dominantly inherited genetically AD mutation05269395
    Display full size

    AD: Alzheimer’s disease; Aβ: amyloid-beta; APP: Aβ precursor protein; MCI: mild cognitive; EOAD: early-onset AD; TREM-2: triggering receptor expressed on myeloid cell 2

    Non-pharmacological interventions

    These interventions include exercise, diet, cognitive training, sleep-related intervention, neurostimulation (with devices), combination therapy, and others (such as environmental enrichment) based on Common Alzheimer’s Disease Research Ontology (CADRO).

    Exercise, diet, cognitive training, and other

    Aerobic exercise for 26 weeks increased glucose metabolism and improved executive function (NCT 02384993) [62]. Social dance movement and exercise with treadmill walking for people with MCI or early stage of AD and other dementia significantly improved multiple brain network characteristics, global efficiency, modularity, quality of life, and balance (NCT 03333837) [63].

    Effects of intermittent calorie restriction on CSF biomarkers related to AD were studied in women with insulin resistance. The result showed a significant reduction of the CSF Aβ42/Aβ40 ratio and reduction of tau (NCT 02460783). Alternative diet therapy includes the nutraceutical huperzine A, vitamin D, omega-3 fatty acids, and probiotics, and has shown benefit in cognitive functions [11, 64]. Nutraceutical huperzine A has shown benefits in improving cognitive function, memory function, and daily living activities by reducing oxidative stress [65]. Clinical trials with omega-3 fatty acids supplements have shown benefits in improving memory probably in link to its cardiovascular benefits in subjects with MCI [11, 66]. Randomized double-blind controlled clinical trials and their meta-analysis showed that probiotics improved cognitive performance and metabolic profiles in AD possibly by reducing intestinal inflammation and oxidative stress [64, 67]. The current early phase 1 triple-masked clinical trial (NCT06181513) is underway to investigate whether giving probiotics (20 million CFU of Lactobacillus paracasei, Lactobacillus plantarum, Lactobacillus rhamnosus, Lactobacillus helveticus, Bifidobacterium breve) to patients with mild AD reduces neuroinflammation, improves cognitive function and modify neurophysiological measures compared to placebo group.

    Brain stimulation with a computer program showed a trend to increase cognitive function and reduce depression in mild to moderate AD in a terminated trial (due to COVID-19) (NCT 03656107) [68]. Behaviorally, play intervention to meet the unmet needs of belonging, esteem, and self-actualization seemed to increase cognitive assessment and memory score in AD (NCT 02846415). Finally, physical and social environmental modification such as ecological therapy with gardening helped to reduce behavioral disorders in moderate to severe AD (NCT 04555616, NCT 02462291).

    Neurostimulation devices

    AD progression from the hippocampus to other parts of the brain disrupts circuitry-level connection (e.g., [69]); that finding led to early clinical trials for inducing gamma-frequency or brain training to promote the connection among brain areas, and cognitive functions. Gamma oscillations using transcranial alternating current stimulation (tACS) at 40 Hz increased blood perfusion in bilateral temporal lobes in mild to moderate AD patients based on an arterial spin labeling study using MRI ([70]; NCT03290326, and NCT 03412604). Similarly, investigators sought to replicate the finding from the AD mouse model study that gamma stimulation with light flicker (gamma entrainment using sensory stimulus or GENUS) reduced phosphorylated tau and amyloid with combined visual and auditory GENUS in the medial prefrontal cortex [71]. A clinical trial (NCT03543878) was designed to entrain human brain activities by exposure to the same gamma oscillation frequency with Flicker devices, licensed by Cognito Therapeutics, in the MCI stage of AD patients; however, the results on AD biomarkers and cognitive behavior scores are not available. On the other hand, transcranial direct current stimulation (tDCS) improved cognition and behavior, with a significant interaction between default mode network (DMN) and salient network and functional connectivity changes in a polarity-dependent effect [72].

    Clinical trials for the neuropsychiatric symptoms of dementia

    Among the few clinical trials targeting non-cognitive symptoms, there are several drugs that show improvement or are FDA-approved (Table 5). Methylphenidate, a dopamine transporter blocker that increases dopamine levels, seems to show mild improvement in apathy, the most prevalent disorder among the neuropsychiatric symptoms of AD [73]. Pimavanserin (Nuplazid), an inverse agonist and antagonist of 5-HT2A in serotonin neurotransmission, shows promising reduction of psychosis. Lastly, lamborexant (Dayvigo) and suvorexant (Belsomra, MK-4305) antagonizing the orexin receptor are approved for insomnia in mild to moderate AD.

    Drugs targeting non-cognitive symptoms of Alzheimer’s disease

    Drug nameTarget and rationaleStatus on clinical trial
    (start date–end date)
    AD stageResultsClinical trial identifier (NCT number)
    MethylphenidateBlock DAT. And increase the dopamine levelCompleted phase 3, (2016–2020)Apathy in ADMild improvement of apathy, measured at 6 months compared to placebo02346201
    PrazosinAntagonist of alpha1-adrenergic receptorCompleted (2018–2022)Moderate to severe disruptive behaviorNon-significant improvement of agitation03710642
    Pimavanserin (Nuplazid)Inverse agonist and antagonist of 5-HT2A in neurotransmissionCompleted phase 2 (2017–2019)Probable AD who has symptoms of agitation and aggression(1) Promising reduction of psychosis score in AD, also see [74] and (2) delay in psychosis relapse (p < 0.05) and a lower risk of relapse with the continuation of the drug [75]03118947
    03325556
    ITI-007 (Lumateperone)5-HT2A serotonin receptor antagonistTerminated phase 3 (2019) due to pre-specified interim analysis indicating futilityProbably ADTerminated02817906
    Lemborexant (Dayvigo)Orexin receptor antagonistCompleted (2016–2020)Mild-moderateApproved for insomnia, lower nighttime activity, and improved circadian rhythmicity03001557
    Suvorexant (Belsomra, MK-4305)Orexin receptor antagonistCompleted (2016–2018)Mild-moderateApproved for insomnia in AD, lengthened total night sleep time02750306
    Display full size

    AD: Alzheimer’s disease; DAT: dopamine transporter; 5-HT2A: 5-hydroxytryptamine 2A

    Conclusions

    Considering that tau proteins correlate with AD more closely than amyloid proteins and participate in the spreading of the disease [51], current multiple ongoing anti-tau immunotherapies are working on improving safety and efficacy and might provide a cheaper affordable, and more efficacious approach in the future, compared to currently approved anti-amyloid therapy against AD. Further, the interplay between Aβ and tau pathology remains to be known, and targeting both pathways could be promising for development of effective therapeutic interventions.

    Future approaches will be first, to improve the drug delivery efficiency through the blood-brain barrier, which will help to reduce the therapeutic dose and reduce side effects (e.g., [10]), and second, to discover therapeutic agents to slow down the decline of several cognitive domains including memory [11]. Further, a precision medicine approach will allow different biomarkers to target the heterogeneity in the pattern of cognitive deficits and presentation in any individual [76]. An equipotential model of AD beyond the amyloid model will warrant specific “precision” treatment to AD, targeting pathology other than amyloid or interacting with tau and amyloid pathology.

    It takes 10–20 years for AD to manifest itself (e.g., [5, 69]). The best approach and current focus of attention would be to predict AD onset as early as possible through the discovery of biomarkers, and then to prevent AD from happening early on or delay the progression of AD, for example, through good diet, aerobic exercise, cognitive stimulation, and enough good quality of sleep and reduction of inflammation (e.g., reviews [10, 28]). For example, various forms of P-tau provide non-invasive cost-effective prediction as biomarkers [50].

    Further, disease-modifying treatment for AD targeting tau, neuroinflammation and neuroprotective mechanisms, immunotherapies, and repurposing existing drugs are under investigation. Combination therapies targeting multiple pathways or multiple aspects of the same pathway may be the key to effective AD treatment and “precision” individualized medicine.

    Abbreviations

    5-HT6:

    5-hydroxytryptamine 6

    AD:

    Alzheimer’s disease

    APOE:

    apolipoprotein E

    Aβ:

    amyloid-beta

    CDR-SB:

    clinical dementia rating-sum of boxes

    CSF:

    cerebral spinal fluid

    MAO:

    monoamine oxidase

    MCI:

    mild cognitive impairment

    NMDA:

    N-methyl-D-aspartic acid

    Declarations

    Acknowledgments

    The authors would like to thank Dr. Lisa Glickstein (Regis College, Weston, MA) for editing the manuscript.

    Author contributions

    DX: Conceptualization, Funding acquisition, Investigation, Writing—original draft, Writing—review & editing. CZ: Conceptualization, Funding acquisition, Writing—review & editing. All authors read and approved the submitted version.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    This work is supported by a Sabbatical Grant (2023) from Regis College, Weston, USA 02493, and a grant from the Chinese Institutes for Medical Research, Beijing [CX23YZ03]. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

    Copyright

    © The Author(s) 2024.

    References

    Colom-Cadena M, Spires-Jones T, Zetterberg H, Blennow K, Caggiano A, DeKosky ST, et al.; Synaptic Health Endpoints Working Group. The clinical promise of biomarkers of synapse damage or loss in Alzheimer's disease. Alzheimers Res Ther. 2020;12:21. [DOI] [PubMed] [PMC]
    Breijyeh Z, Karaman R. Comprehensive Review on Alzheimer's Disease: Causes and Treatment. Molecules. 2020;25:5789. [DOI] [PubMed] [PMC]
    Collaborators G2CoD. Global burden of 288 causes of death and life expectancy decomposition in 204 countries and territories and 811 subnational locations, 1990-2021: a systematic analysis for the Global Burden of Disease Study 2021. Lancet. 2024;403:210032. [DOI] [PubMed] [PMC]
    Lane CA, Hardy J, Schott JM. Alzheimer’s disease. Eur J Neurol. 2018;25:5970. [DOI] [PubMed]
    Ballard C, Gauthier S, Corbett A, Brayne C, Aarsland D, Jones E. Alzheimer’s disease. Lancet. 2011;377:101931. [DOI] [PubMed]
    Giri M, Zhang M, Lü Y. Genes associated with Alzheimer's disease: an overview and current status. Clin Interv Aging. 2016;11:66581. [DOI] [PubMed] [PMC]
    Karch CM, Goate AM. Alzheimer’s disease risk genes and mechanisms of disease pathogenesis. Biol Psychiatry. 2015;77:4351. [DOI] [PubMed] [PMC]
    Lanfranco MF, Ng CA, Rebeck GW. ApoE Lipidation as a Therapeutic Target in Alzheimer’s Disease. Int J Mol Sci. 2020;21:6336. [DOI] [PubMed] [PMC]
    Athar T, Balushi KA, Khan SA. Recent advances on drug development and emerging therapeutic agents for Alzheimer’s disease. Mol Biol Rep. 2021;48:56295645. [DOI] [PubMed] [PMC]
    Passeri E, Elkhoury K, Morsink M, Broersen K, Linder M, Tamayol A, et al. Alzheimer's Disease: Treatment Strategies and Their Limitations. Int J Mol Sci. 2022;23:13954. [DOI] [PubMed] [PMC]
    Weller J, Budson A. Current understanding of Alzheimer’s disease diagnosis and treatment. F1000Res. 2018;7:F1000 Faculty Rev1161. [DOI] [PubMed] [PMC]
    Arvanitakis Z, Shah RC, Bennett DA. Diagnosis and Management of Dementia: Review. JAMA. 2019;322:158999. [DOI] [PubMed] [PMC]
    Nguyen K, Hoffman H, Chakkamparambil B, Grossberg GT. Evaluation of rivastigmine in Alzheimer’s disease. Neurodegener Dis Manag. 2021;11:3548. [DOI] [PubMed]
    Cummings J, Osse AML, Cammann D, Powell J, Chen J. Anti-Amyloid Monoclonal Antibodies for the Treatment of Alzheimer’s Disease. BioDrugs. 2024;38:522. [DOI] [PubMed] [PMC]
    Perneczky R, Dom G, Chan A, Falkai P, Bassetti C. Anti-amyloid antibody treatments for Alzheimer’s disease. Eur J Neurol. 2024;31:e16049. [DOI]
    Heneka MT, Carson MJ, Khoury JE, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388405. [DOI] [PubMed] [PMC]
    Sun X, Li L, Dong Q, Zhu J, Huang Y, Hou S, et al. Rutin prevents tau pathology and neuroinflammation in a mouse model of Alzheimer’s disease. J Neuroinflammation. 2021;18:131. [DOI] [PubMed] [PMC]
    Heneka MT. ApoE4 makes microglia trem2 bling. Neuron. 2023;111:1424. [DOI] [PubMed]
    Macdonald R, Barnes K, Hastings C, Mortiboys H. Mitochondrial abnormalities in Parkinson’s disease and Alzheimer’s disease: can mitochondria be targeted therapeutically? Biochem Soc Trans. 2018;46:891909. [DOI] [PubMed]
    Wunderlich G, Blahova Z, Garcia M, Jessen F. Efficacy and safety of the novel GlyT1 inhibitor BI 425809 in Alzheimer’s dementia: a randomized controlled trial. Alzheimers Res Ther. 2023;15:24. [DOI] [PubMed] [PMC]
    Hajjar I, Okafor M, McDaniel D, Obideen M, Dee E, Shokouhi M, et al. Effects of Candesartan vs Lisinopril on Neurocognitive Function in Older Adults With Executive Mild Cognitive Impairment: A Randomized Clinical Trial. JAMA Netw Open. 2020;3:e2012252. [DOI] [PubMed] [PMC]
    Chase TN, Farlow MR, Clarence-Smith K. Donepezil Plus Solifenacin (CPC-201) Treatment for Alzheimer's Disease. Neurotherapeutics. 2017;14:40516. [DOI] [PubMed] [PMC]
    Ostrowitzki S, Bittner T, Sink KM, Mackey H, Rabe C, Honig LS, et al. Evaluating the Safety and Efficacy of Crenezumab vs Placebo in Adults With Early Alzheimer Disease: Two Phase 3 Randomized Placebo-Controlled Trials. JAMA Neurol. 2022;79:11131121. [DOI] [PubMed] [PMC]
    Yu HJ, Dickson SP, Wang P, Chiu M, Huang C, Chang C, et al. Safety, tolerability, immunogenicity, and efficacy of UB-311 in participants with mild Alzheimer’s disease: a randomised, double-blind, placebo-controlled, phase 2a study. EBioMedicine. 2023;94:104665. [DOI] [PubMed] [PMC]
    Lon HK, Mendonca N, Goss S, Othman AA, Locke C, Jin Z, et al. Pharmacokinetics, Safety, Tolerability, and Pharmacodynamics of Alicapistat, a Selective Inhibitor of Human Calpains 1 and 2 for the Treatment of Alzheimer Disease: An Overview of Phase 1 Studies. Clin Pharmacol Drug Dev. 2019;8:290303. [DOI] [PubMed]
    Novak P, Schmidt R, Kontsekova E, Zilka N, Kovacech B, Skrabana R, et al. Safety and immunogenicity of the tau vaccine AADvac1 in patients with Alzheimer’s disease: a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet Neurol. 2017;16:12334. [DOI]
    Novak P, Kovacech B, Katina S, Schmidt R, Scheltens P, Kontsekova E, et al. ADAMANT: a placebo-controlled randomized phase 2 study of AADvac1, an active immunotherapy against pathological tau in Alzheimer’s disease. Nat Aging. 2021;1:52134. [DOI] [PubMed]
    Cummings JL, Tong G, Ballard C. Treatment Combinations for Alzheimer’s Disease: Current and Future Pharmacotherapy Options. J Alzheimers Dis. 2019;67:77994. [DOI] [PubMed] [PMC]
    Malar DS, Thitilertdecha P, Ruckvongacheep KS, Brimson S, Tencomnao T, Brimson JM. Targeting Sigma Receptors for the Treatment of Neurodegenerative and Neurodevelopmental Disorders. CNS Drugs. 2023;37:399440. [DOI] [PubMed] [PMC]
    Hampel H, Williams C, Etcheto A, Goodsaid F, Parmentier F, Sallantin J, et al. A precision medicine framework using artificial intelligence for the identification and confirmation of genomic biomarkers of response to an Alzheimer’s disease therapy: Analysis of the blarcamesine (ANAVEX2-73) Phase 2a clinical study. Alzheimers Dement (N Y). 2020;6:e12013. [DOI] [PubMed] [PMC]
    Wessels AM, Lines C, Stern RA, Kost J, Voss T, Mozley LH, et al. Cognitive outcomes in trials of two BACE inhibitors in Alzheimer’s disease. Alzheimers Dement. 2020;16:148392. [DOI] [PubMed]
    Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid beta-peptide. Nat Rev Mol Cell Biol. 2007;8:10112. [DOI] [PubMed]
    Pluta R, Barcikowska M, Misicka A, Lipkowski AW, Spisacka S, Januszewski S. Ischemic rats as a model in the study of the neurobiological role of human beta-amyloid peptide. Neuroreport. 1999;10:36159. [DOI] [PubMed]
    Yadollahikhales G, Rojas JC. Anti-Amyloid Immunotherapies for Alzheimer’s Disease: A 2023 Clinical Update. Neurotherapeutics. 2023;20:914931. [DOI] [PubMed] [PMC]
    Vandenberghe R, Rinne JO, Boada M, Katayama S, Scheltens P, Vellas B, et al.; Bapineuzumab 3000 and 3001 Clinical Study Investigators. Bapineuzumab for mild to moderate Alzheimer’s disease in two global, randomized, phase 3 trials. Alzheimers Res Ther. 2016;8:18. [DOI] [PubMed] [PMC]
    Honig LS, Vellas B, Woodward M, Boada M, Bullock R, Borrie M, et al. Trial of Solanezumab for Mild Dementia Due to Alzheimer’s Disease. N Engl J Med. 2018;378:32130. [DOI]
    Asher S, Priefer R. Alzheimer’s disease failed clinical trials. Life Sci. 2022;306:120861. [DOI] [PubMed]
    Tagliapietra, M. Aducanumab for the treatment of Alzheimer’s disease. Drugs Today (Barc). 2022;58:46577. [DOI] [PubMed]
    Jorfi M, Maaser-Hecker A, Tanzi RE. The neuroimmune axis of Alzheimer’s disease. Genome Med. 2023;15:6. [DOI] [PubMed] [PMC]
    Sims JR, Zimmer JA, Evans CD, Lu M, Ardayfio P, Sparks J, et al.; TRAILBLAZER-ALZ 2 Investigators. Donanemab in Early Symptomatic Alzheimer Disease: The TRAILBLAZER-ALZ 2 Randomized Clinical Trial. JAMA. 2023;330:51227. [DOI] [PubMed] [PMC]
    Pontecorvo MJ, Lu M, Burnham SC, Schade AE, Dage JL, Shcherbinin S, et al. Association of Donanemab Treatment With Exploratory Plasma Biomarkers in Early Symptomatic Alzheimer Disease: A Secondary Analysis of the TRAILBLAZER-ALZ Randomized Clinical Trial. JAMA Neurol. 2022;79:12509. [DOI] [PubMed] [PMC]
    Reardon, S. Alzheimer’s drug donanemab helps most when taken at earliest disease stage, study finds. Nature. 2023;619:6823. [DOI]
    van Dyck CH, Sabbagh M, Cohen S. Lecanemab in Early Alzheimer’s Disease. N Engl J Med. 2023;388:921. [DOI] [PubMed]
    Reiman EM, Pruzin JJ, Rios-Romenets S, Brown C, Giraldo M, Acosta-Baena N, et al.; API ADAD Colombia Trial Group. A public resource of baseline data from the Alzheimer’s Prevention Initiative Autosomal-Dominant Alzheimer’s Disease Trial. Alzheimers Dement. 2023;19:193846. [DOI] [PubMed] [PMC]
    Tariot PN, Lopera F, Langbaum JB, Thomas RG, Hendrix S, Schneider LS, et al. The Alzheimer’s Prevention Initiative Autosomal-Dominant Alzheimer’s Disease Trial: A study of crenezumab versus placebo in preclinical PSEN1 E280A mutation carriers to evaluate efficacy and safety in the treatment of autosomal-dominant Alzheimer’s disease, including a placebo-treated noncarrier cohort. Alzheimers Dement (N Y). 2018;4:15060. [DOI] [PubMed] [PMC]
    Robinson SR, Bishop GM, Lee HG, Münch G. Lessons from the AN 1792 Alzheimer vaccine: lest we forget. Neurobiol Aging. 2004;25:60915. [DOI] [PubMed]
    Wang CY, Wang P, Chiu M, Finstad CL, Lin F, Lynn S, et al. UB-311, a novel UBITh® amyloid β peptide vaccine for mild Alzheimer’s disease. Alzheimers Dement (N Y). 2017;3:26272. [DOI] [PubMed] [PMC]
    Cullen NC, Novak P, Tosun D, Kovacech B, Hanes J, Kontsekova E, et al. Efficacy assessment of an active tau immunotherapy in Alzheimer’s disease patients with amyloid and tau pathology: a post hoc analysis of the “ADAMANT” randomised, placebo-controlled, double-blind, multi-centre, phase 2 clinical trial. EBioMedicine. 2024;99:104923. [DOI] [PubMed] [PMC]
    Krishnadas N, Doré V, Robertson JS, Ward L, Fowler C, Masters CL, et al. Rates of regional tau accumulation in ageing and across the Alzheimer’s disease continuum: an AIBL 18F-MK6240 PET study. EBioMedicine. 2023;88:104450. [DOI] [PubMed] [PMC]
    Ossenkoppele R, van der Kant R, Hansson O. Tau biomarkers in Alzheimer’s disease: towards implementation in clinical practice and trials. Lancet Neurol. 2022;21:72634. [DOI] [PubMed]
    Sandusky-Beltran LA, Sigurdsson EM. Tau immunotherapies: Lessons learned, current status and future considerations. Neuropharmacology. 2020;175:108104. [DOI] [PubMed] [PMC]
    Teng E, Manser PT, Pickthorn K, Brunstein F, Blendstrup M, Bohorquez SS, et al.; Tauriel Investigators. Safety and Efficacy of Semorinemab in Individuals With Prodromal to Mild Alzheimer Disease: A Randomized Clinical Trial. JAMA Neurol. 2022;79:75867. [DOI] [PubMed] [PMC]
    Colom-Cadena M, Davies C, Sirisi S, Lee J-E, Simzer EM, Tzioras M, et al. Synaptic oligomeric tau in Alzheimer’s disease - A potential culprit in the spread of tau pathology through the brain. Neuron. 2023;11:217083.e6. [DOI]
    Matthews DC, Mao X, Dowd K, Tsakanikas D, Jiang CS, Meuser C, et al. Riluzole, a glutamate modulator, slows cerebral glucose metabolism decline in patients with Alzheimer's disease. Brain. 2021;144:374255. [DOI] [PubMed] [PMC]
    Matthews DC, Ritter A, Thomas RG, Andrews RD, Lukic AS, Revta C, et al. Rasagiline effects on glucose metabolism, cognition, and tau in Alzheimer’s dementia. Alzheimers Dement (N Y). 2021;7:e12106. [DOI] [PubMed] [PMC]
    Ruthirakuhan MT, Herrmann N, Gallagher D, Andreazza AC, Kiss A, Verhoeff NPLG, et al. Investigating the safety and efficacy of nabilone for the treatment of agitation in patients with moderate-to-severe Alzheimer’s disease: Study protocol for a cross-over randomized controlled trial. Contemp Clin Trials Commun. 2019;15:100385. [DOI] [PubMed] [PMC]
    Ruthirakuhan M, Herrmann N, Andreazza AC, Verhoeff NPLG, Gallagher D, Black SE, et al. Agitation, Oxidative Stress, and Cytokines in Alzheimer Disease: Biomarker Analyses From a Clinical Trial With Nabilone for Agitation. J Geriatr Psychiatry Neurol. 2020;33:17584. [DOI] [PubMed]
    Nirogi R, Goyal VK, Benade V, Subramanian R, Ravula J, Jetta S, et al. Effect of Concurrent Use of Memantine on the Efficacy of Masupirdine (SUVN-502): A Post Hoc Analysis of a Phase 2 Randomized Placebo-Controlled Study. Neurol Ther. 2022;11:158394. [DOI] [PubMed] [PMC]
    Nirogi R, Ieni J, Goyal VK, Ravula J, Jetta S, Shinde A, et al. Effect of masupirdine (SUVN-502) on cognition in patients with moderate Alzheimer’s disease: A randomized, double-blind, phase 2, proof-of-concept study. Alzheimers Dement (N Y). 2022;8:e12307. [DOI] [PubMed] [PMC]
    Hori Y, Takeda S, Cho H, Wegmann S, Shoup TM, Takahashi K, et al. A Food and Drug Administration-approved asthma therapeutic agent impacts amyloid β in the brain in a transgenic model of Alzheimer disease. J Biol Chem. 2015;290:196678. [DOI] [PubMed] [PMC]
    Busche MA, Hyman BT. Synergy between amyloid-β and tau in Alzheimer’s disease. Nat Neurosci. 2020:23:11831193. [DOI]
    Gaitán JM, Boots EA, Dougherty RJ, Oh JM, Ma Y, Edwards DF, et al. Brain Glucose Metabolism, Cognition, and Cardiorespiratory Fitness Following Exercise Training in Adults at Risk for Alzheimer’s Disease. Brain Plast. 2019;5:8395. [DOI] [PubMed] [PMC]
    Thumuluri D, Lyday R, Babcock P, Ip EH, Kraft RA, Laurienti PJ, et al. Improvisational Movement to Improve Quality of Life in Older Adults With Early-Stage Dementia: A Pilot Study. Front Sports Act Living. 2022;3:796101. [DOI] [PubMed] [PMC]
    Thakkar A, Vora A, Kaur G, Akhtar J. Dysbiosis and Alzheimer’s disease: role of probiotics, prebiotics and synbiotics. Naunyn Schmiedebergs Arch Pharmacol. 2023;396:291123. [DOI] [PubMed]
    Sen D, Rathee S, Pandey V, Jain SK, Patil UK. Comprehensive Insights into Pathophysiology of Alzheimer’s Disease: Herbal Approaches for Mitigating Neurodegeneration. Curr Alzheimer Res. Forthcoming 2024. [DOI] [PubMed]
    Jia X, Kohli P, Virani SS. Omega-3 Fatty Acid and Cardiovascular Outcomes: Insights From Recent Clinical Trials. Curr Atheroscler Rep. 2019;10;21:1. [DOI] [PubMed]
    Den H, Dong X, Chen M, Zou Z. Efficacy of probiotics on cognition, and biomarkers of inflammation and oxidative stress in adults with Alzheimer’s disease or mild cognitive impairment — a meta-analysis of randomized controlled trials. Aging (Albany NY). 2020;12:401039. [DOI] [PubMed] [PMC]
    Beishon L, Evley R, Panerai RB, Subramaniam H, Mukaetova-Ladinska E, Robinson T, et al. Effects of brain training on brain blood flow (The Cognition and Flow Study-CogFlowS): protocol for a feasibility randomised controlled trial of cognitive training in dementia. BMJ Open. 2019;9:e027817. [DOI] [PubMed] [PMC]
    Xiao D, Wang K, Theriault L, Charbel E; Alzheimer’s Disease Neuroimaging Initiative. White matter integrity and key structures affected in Alzheimer’s disease characterized by diffusion tensor imaging. Eur J Neurosci. 2022;56:531931. [DOI]
    Sprugnoli G, Munsch F, Cappon D, Paciorek R, Macone J, Connor A, et al. Impact of multisession 40Hz tACS on hippocampal perfusion in patients with Alzheimer's disease. Alzheimers Res Ther. 2021;13:203. [DOI] [PubMed] [PMC]
    Martorell AJ, Paulson AL, Suk H, Abdurrob F, Drummond GT, Guan W, et al. Multi-sensory Gamma Stimulation Ameliorates Alzheimer’s-Associated Pathology and Improves Cognition. Cell. 2019;177:256271.e22. [DOI] [PubMed] [PMC]
    Pini L, Pizzini FB, Boscolo-Galazzo I, Ferrari C, Galluzzi S, Cotelli M, et al. Brain network modulation in Alzheimer’s and frontotemporal dementia with transcranial electrical stimulation. Neurobiol Aging. 2022;111:2434. [DOI]
    Zhao Q, Tan L, Wang H, Jiang T, Tan M, Tan L, et al. The prevalence of neuropsychiatric symptoms in Alzheimer's disease: Systematic review and meta-analysis. J Affect Disord. 2016;190:26471. [DOI] [PubMed]
    Ismail Z, Creese B, Aarsland D, Kales HC, Lyketsos CG, Sweet RA, et al. Psychosis in Alzheimer disease - mechanisms, genetics and therapeutic opportunities. Nat Rev Neurol. 2022;18:13144. [DOI] [PubMed] [PMC]
    Tariot PN, Cummings JL, Soto-Martin ME, Ballard C, Erten-Lyons D, Sultzer DL, et al. Trial of Pimavanserin in Dementia-Related Psychosis. N Engl J Med. 2021;385:30919. [DOI] [PubMed]
    Bondi MW, Edmonds EC, Salmon DP. Alzheimer’s Disease: Past, Present, and Future. J Int Neuropsychol Soc. 2017;23:81831. [DOI] [PubMed] [PMC]