Abstract
Drug repositioning and repurposing can enhance traditional drug development efforts and could accelerate the identification of new treatments for individuals with Alzheimer disease (AD) dementia and mild cognitive impairment. Transcriptional profiling offers a new and highly efficient approach to the identification of novel candidates for repositioning and repurposing. In the future, novel AD transcriptional signatures from cells isolated at early stages of disease, or from human neurons or microglia that carry mutations that increase the risk of AD, might be used as probes to identify additional candidate drugs. Phase II trials assessing repurposed agents must consider the best target population for a specific candidate therapy as well as the mechanism of action of the treatment. In this Review, we highlight promising compounds to prioritize for clinical trials in individuals with AD, and discuss the value of Delphi consensus methodology and evidence-based reviews to inform this prioritization process. We also describe emerging work, focusing on the potential value of transcript signatures as a cost-effective approach to the identification of novel candidates for repositioning.
The growing global health challenge posed by dementia needs to be addressed. Currently, more than 40 million people have Alzheimer disease (AD) worldwide, and this number is expected to increase to more than 100 million by 2050 (REF.1). In addition, estimates indicate that at least 15% of people aged 60 years or above have mild cognitive impairment (MCI) and that 8–15% of these individuals will progress from MCI to dementia each year, most commonly to AD2. AD is a devastating, progressive neurodegenerative disease that has a massive personal and financial impact on individuals, families and society. The estimated annual cost of dementia worldwide is US$818 billion, which is predicted to increase to US$1 trillion within this decade1. In the past 20 years only two new pharmacological therapies have become available for the treatment of AD. One of the treatments, memantine, has been licensed for the treatment of AD globally, whereas the other, oligomannate, is only licensed in China. Importantly, no pharmacological treatments have been licensed for use in individuals with MCI.
The core pathological substrates of AD in the brain are amyloid plaques and neurofibrillary tangles; the latter involve the hyperphosphorylation of tau3. The importance of other potential mechanisms, including neuroinflammation, protein misfolding, mitochondrial dysfunction and clearance of abnormal proteins, in the pathophysiology of AD has become increasingly apparent4. Despite a number of controversies regarding the role of amyloid in the pathogenesis of AD, including the question of whether neuronal death is driven by amyloid plaques or soluble amyloid and oligomers5, the vast majority of treatments evaluated in clinical trials have focused on amyloid-related targets. The past decade has seen a number of high profile unsuccessful randomized clinical trials (RCTs) of amyloid-focused treatments, for example the anti-amyloid immunotherapy solanezumab6 and the β-secretase inhibitor verubecestat7. A recent review of the NIH clinical trial registry identified only 29 pharmacological or biological treatments in ongoing phase II or phase III trials for disease modification in AD or MCI8. This number is 40-fold less than the number of ongoing RCTs in cancer8, and the number of RCTs of disease-modifying therapies for AD has not substantially increased since 2012 (REF.9). Despite the enormous potential value of an effective disease-modifying therapy for AD or MCI, this area of research is considered to be high risk by the pharmaceutical industry, particularly as a result of low clinical trial success rates, and a number of global pharmaceutical companies have withdrawn investment from this therapeutic area10. Multiple factors could be responsible for the failed trials of disease-modifying therapies for AD; for example, the use of suboptimal treatments and targets, a narrow range of targets and methodological issues with the trials (BOX 1). Furthermore, owing to the low sensitivity of clinical and neuropsychological outcome measures, nearly 500 participants per treatment arm are needed for adequately powered phase II trials in individuals with MCI, which means that many phase II trials in individuals with this condition are significantly under-powered and the results are difficult to interpret11.
Box 1 |. Potential reasons for failed RCTs of AD therapies.
Therapeutics and targets
The vast majority of trials have focused on amyloid targets, resulting in a lack of breadth.
There is uncertainty regarding the specific disease mechanisms related to different amyloid species.
Some therapeutics show poor brain penetration.
Reducing amyloid deposition alone might not be sufficient to induce disease-modifying changes.
There has been only limited use of target engagement biomarkers in phase II studies to inform phase III studies.
Trial design
Many trials might have been performed in individuals with Alzheimer disease (AD) that has progressed too far for therapies to have a disease-modifying effect; an increased focus on preclinical AD and at-risk groups has been seen in more recent trials.
The results of phase II trials have been interpreted in an overly optimistic manner, leading to the progression of some compounds to larger trials that might not have been warranted.
Populations that are appropriately enriched for core AD pathologies have only been included in more recent trials.
The neuropsychology measures used in trials can have a poor sensitivity to change; this insensitivity is a particular issue in phase II trials, which have usually been under-powered to detect changes in neuropsychology and clinical outcomes.
RCT, randomized clinical trial.
The results from trials of the amyloid-targeting antibody aducanumab are emerging. In one of the two completed phase III trials, participants receiving aducanumab showed a statistically significant improvement in cognition and function compared with participants receiving placebo, particularly in the groups of participants carrying APOE ε4 (REF.12). The data from the other phase III trial are less clear, although some indication of benefit in participants exposed to higher doses was reported12. The results of these trials are not yet fully in the public domain and have not been subjected to peer review, so interpretation needs to be cautious. Therapies that focus on other key treatment targets such as tau and neuroinflammation are at an earlier stage of development than aducanumab, but the preclinical data are promising13. These encouraging results might have a positive impact on AD drug discovery, for example, by attracting increased investment from the pharmaceutical industry. However, complementing traditional drug discovery with a broader range of approaches, such as drug repositioning and repurposing, will maximize drug development efforts. We used a systematic review of the literature and a Delphi consensus approach to highlight existing compounds that we feel should be prioritized for clinical trials in individuals with AD. In this Review, we present the results of that Delphi consensus and describe the evidence underlying the consensus prioritization. We then describe emerging work, focusing on the potential value of transcript signatures as a cost-effective approach to identifying novel candidates for repositioning.
Drug repositioning and repurposing
Drug repositioning occurs within the biopharma industry during drug development and refers to the development of an agent for an indication other than the indication it was originally intended for. This new indication is prioritized during the development process and before approval. By contrast, drug repurposing is defined as “the application of established drug compounds to new therapeutic indications”14 and offers a route to drug development that is accessible to academic institutions, government and research council programmes, charities and not-for-profit organizations, thus complementing the work of pharmaceutical and biotechnology companies. Repositioning and repurposing offer an attractive way of enhancing traditional drug development and accelerating the arrival of new treatments for AD dementia and MCI in the clinic. Phase II trials assessing repurposed agents must consider the best target population for a specific candidate therapy as well as the mechanism of action of the treatment.
Drug repurposing has enabled the identification of successful therapies for many diseases including cancer15 and Parkinson disease (PD)16. One important advantage of this approach is that the safety of the candidate compound has already been established, which removes the need for further preclinical safety testing, chemical optimization or toxicology studies, and thus substantially reduces the time and cost involved in progressing the potential treatment into clinical trials. Marketed drugs are likely to have a reasonable safety database derived from previous registrational programmes, postmarketing experience and safety surveillance. In many cases, understanding this safety profile offers a solid ‘freedom to operate’ when repurposing the drug in a relatively fragile population, such as individuals with AD. Drug repurposing might also offer the further key advantage of bypassing the early preclinical, phase II and even phase IIa trials, all of which are time consuming and represent periods of relatively high drug attrition. In addition, many of the costs of drug development that are not always readily recognized, such as those associated with formulation optimization, manufacturing development and drug–drug interaction studies, have been addressed by the biopharmaceutical company that originally developed the drug. The estimated cost of developing a drug to the point of approval is US$5.6 billion17, but these extreme costs can be lower in programmes that focus on repurposed agents. Furthermore, for repurposed agents, clinical evidence of potential efficacy can be derived from existing pathophysiological observations, epidemiological cohort studies, open-treatment studies and preliminary clinical trials. This clinical information provides an important added dimension to the available evidence, particularly given the limitations of animal models.
Candidates for drug repurposing can be selected via a number of different routes, one of which is the use of large datasets to detect drug-associated patient outcomes that would otherwise have not been identified18. An alternative route is hypothesis-driven repurposing, which combines information about the disease of interest and the properties and targets of existing drugs for other conditions to identify potential candidates9. Similarly, high-throughput screening using in vitro models designed to assess the effects of compounds on known target mechanisms, such as amyloid toxicity, can be used19. A novel method is the use of disease-associated transcriptional signatures as a tool for identifying candidate therapies20.
Another approach is to combine several of the above sources of information by manually reviewing the existing literature to identify candidates for repurposing. The challenge is that the kind of evidence available often varies among different compounds; for example, strong in vitro or in vivo evidence might exist for some candidates, whereas strong epidemiological evidence might exist for others. In addition, any identified treatment has to also be suitable for the target population, which for AD is older individuals with dementia. One way of addressing this challenge is to combine systematic review of the evidence with rigorous expert interpretation and consensus using methodologies such as the Delphi consensus approach, which is a standardized approach to achieving expert consensus based on a standardized review of the evidence and serial re-rating of priorities by a panel of experts.
The Delphi consensus process
In writing this Review we combined available evidence from the repurposing routes described in the previous section with the aim of identifying the best candidate compounds for the treatment of AD or MCI. This process involved a comprehensive assessment of the published literature, a systematic evaluation of the evidence and a formal Delphi consensus process involving an expert panel. The Delphi panel had 12 members with expertise from the pharmaceutical industry, academia or drug development funding within the charity sector, including the authors of this Review (with the exception of G.W., P.D., A.C. and J.S.) and three additional panel members who represented patient organizations (see Acknowledgements section). Each panel member was asked to nominate up to ten candidate compounds for further consideration. A full systematic review of the literature was prepared for all five candidate compounds that were identified by at least three members of the panel. The members of the panel then ranked these five drug candidates in order of priority on the basis of the strength of evidence. The key factors used for this ranking included the mechanism and efficiency of brain penetration, the safety profile of the compound and whether or not the dosage of the drug used in preclinical studies was equivalent to the safe human dosage. The prioritization ratings of each panel member were shared with the panel at a face-to-face meeting and a second prioritization exercise was undertaken by e-mail. The prioritization was then finalized at a further face-to-face meeting of the panel. This methodology was designed to update the systematic review and Delphi consensus published in 2012 in Nature Reviews Drug Discovery9. As the aim of this second Delphi consensus was to identify new candidate compounds, priority candidates from the 2012 census were excluded, but candidates not prioritized by the 2012 consensus were eligible if new evidence had emerged.
Update on existing priority compounds
The 2012 Delphi consensus9 prioritized five classes of compounds for repurposing as treatments for AD: tetracycline antibiotics, calcium channel blockers, angiotensin receptor blockers (ARBs), glucagon-like peptide 1 (GLP1) analogues and retinoid therapy. With the exception of retinoid therapy, all of the prioritized classes of compounds have now been taken into clinical trials. Trials of the tetracycline antibiotic minocycline21, the calcium channel blocker nilvadipine22 and the ARB losartan23 have been completed and did not find any significant benefits of treatment on the cognition or function of individuals with AD.
Tetracycline antibiotics
The RCT of minocycline21 was a three-arm 24-month trial that compared the effects of either 400 mg minocycline per day, 200 mg minocycline per day or placebo, in a total of 554 participants with mild AD and a Mini Mental Status Examination (MMSE) score of ≥24. The two groups of participants who received the minocycline treatment were combined for the data analysis. In this combined group, the change in mean MMSE score, the primary outcome measure, over 24 months was only 0.1 points less than in the group that received placebo. No difference in the change in ability to perform activities of daily living over the 24 months was detected between the two groups. This was a pragmatic, but well-designed study, and provides a clear negative result, which suggests that further trials of minocycline for the treatment of AD are not warranted.
Calcium channel blockers
Nilvadipine (8 mg per day) was evaluated in an 18-month double-blind RCT in 511 participants, of whom 253 received nilvadipine and 258 received placebo22. The participants were over the age of 50 years and had an MMSE score between 12 and 27, thus meeting the National Institute of Neurological and Communicative Disorders and Stroke — Alzheimer’s Disease and Related Disorders Association (NINCDS-ADRDA) criteria for probable AD24. The primary outcome measure was a change in Alzheimer’s Disease Assessment Scale – Cognitive Subscale (ADAS-Cog) score; however, only a 0.21-point non-significant difference in average ADAS-Cog score was observed between the two treatment groups over 18 months. For context, studies of cholinesterase inhibitors have found differences of >2 points in the ADAS-Cog score between groups of participants receiving treatment and groups of participants receiving placebo25, and this would usually be regarded as the minimum clinically meaningful degree of change26. No benefit of treatment with nilvadipine was detected with the co-primary outcome measure (Clinical Dementia Rating — Sum of Boxes), or on any of the secondary or exploratory outcome measures. This trial was well designed and adequately powered and the absence of any significant differences between groups is clearly a negative result, and plans for further studies of nilvadipine for the treatment of AD have not been reported.
Angiotensin receptor blockers
In a preliminary study, 20 participants with probable AD and essential hypertension were randomly assigned to receive either the ARB telmisartan (ten participants, 40–80 mg per day) or the calcium channel blocker amlodipine (ten participants, 5–10 mg per day) for 6 months27. The group of participants who received telmisartan had increased regional cerebral blood flow in the right supramarginal gyrus, superior parietal lobule, cuneus and lingual gyrus compared with the group of participants that received amlodipine. No differences in cognition were observed between the two groups, but the study was very under-powered for detecting neuropsychological outcomes. More recently, in an RCT of the ARB losartan, 211 participants with mild or moderate AD were randomly assigned to receive either 100 mg losartan or placebo once daily for 12 months23. Preliminary results from the trial were presented at the Clinical Trials on Alzheimer’s Disease (CTAD) conference in 2019. No significant reduction in the rate of cortical atrophy, which was the primary outcome measure, was observed in the participants receiving losartan compared with those receiving placebo, and the other clinical and cognitive outcome measures showed no indication of improvement associated with losartan treatment. Although the trial was under-powered for detecting changes in clinical outcomes, the absence of any trends towards improvement in the treatment group was disappointing23.
Despite these negative clinical trial results, a solid body of in vitro and in vivo work supports the potential utility of ARBs as a treatment for AD28–40. In vitro work has identified multiple effects of centrally acting angiotensin II, including vasoconstriction, mitochondrial dysfunction, inhibition of acetylcholine release, increased production of angiotensin IV and release of inflammatory mediators28–30, that suggest ARBs could be suited to repurposing for AD. Many commonly used ARBs, such as candesartan and losartan, have known blood–brain barrier penetration properties and have been shown to attenuate the central effects of angiotensin II in animal studies31. For example, in one study treatment with the ARB valsartan was associated with reduced amyloid-β (Aβ) aggregation in vitro32, and improvements in behavioural tests of cognitive performance and reductions in amyloid pathology in a mouse model of AD32. In other studies of mouse models of AD, animals treated with ARBs showed reduced brain levels of total amyloid or Aβ aggregation, improvements in cognition and reduced neuroinflammation compared with animals treated with saline33–37. Studies of ARBs in Sprague Dawley rats have produced contradictory results, with some studies finding an ARB-associated decrease in tau phosphorylation and some studies finding an ARB-associated increase in tau phosphorylation38–40.
Some epidemiological evidence also supports the use of ARBs for the treatment of AD. A large 4-year study of the medical records of 800,000 adults aged over 65 years found an almost 50% reduction in incident AD in individuals receiving ARBs compared with individuals receiving other cardiovascular treatments. The ONTARGET trial included 16,000 participants with hypertension, and significantly fewer participants showed a decrease in MMSE score to <18 in the group receiving the ARB telmisartan than in the group receiving the ACE inhibitor ramipril41. However, this finding was not replicated in the parallel TRANSCEND trial in 5,000 participants with hypertension, which compared telmisartan with placebo41, nor in the SCOPE trial in nearly 5,000 participants with hypertension, which compared the ARB candesartan with placebo. However, a subgroup analysis in participants from the SCOPE trial with pretreatment MMSE scores of 24–28 showed a modest benefit of treatment on cognitive ability42.
The overall evidence for the use of ARBs to treat AD is mixed, and the absence of any benefits in the RCT of losartan is disappointing. However, the evidence reviewed in this section focuses on specific treatment mechanisms that are related directly to actions on the renin–angiotensin system. These observations must be interpreted in the context of strong epidemiological evidence indicating that hypertension is a risk factor for AD dementia43 and the results of the recent SPRINT MIND trial, which demonstrated a significant reduction in the risk of MCI and probable AD dementia in participants receiving intensive anti-hypertensive management (target systolic blood pressure <120 mmHg) compared with the usual anti-hypertensive management (target systolic blood pressure <140 mmHg)44. The potential overall benefits of blood pressure reduction for heart and brain health should also be considered. Indeed, RCTs of candesartan and telmisartan in individuals with or at risk of AD are ongoing, and we should not discount ARBs as a potential treatment until the results of these trials are reported45–47.
GLP1 analogues
The emerging evidence base for the use of GLP1 analogues to treat AD is more encouraging than that of the other compounds prioritized by the 2012 Delphi consensus9. GLP1 analogues were prioritized on the basis of several in vivo studies in mouse models of AD that demonstrated an effect of this treatment on amyloid and tau pathologies48–51 as well as oxidative stress, apoptosis, synaptic plasticity and other core neuronal functions49,51–57. More recently, this work was extended by a study of the GLP1 analogue liraglutide58. In this study, treatment of amyloid precursor protein–presenilin 1 (APP–PS1) mice (which carry AD-associated mutations in App and PSEN1) with liraglutide from the age of 2 months attenuated the development of progressive AD-related pathological changes, such as synapse loss, synaptic plasticity and amyloid plaques. Indeed, treatment with liraglutide has consistently been associated with improvements in cognition and memory in animal models of AD58–61.
Three randomized, double-blind, multicentre, placebo-controlled trials examining of the cardiovascular effects of liraglutide or semaglutide also included the development of dementia as an exploratory outcome. A total of 15,820 participants were included in the three trials, and the median follow-up period was 3.6 years. Across the three trials, 15 participants who received a GLP1 analogue and 32 participants who received placebo developed dementia, with an estimated hazard ratio of 0.47 (95% CI 0.25–0.86) in favour of the GLP1 analogue treatment (C.B., unpublished work). This analysis is exploratory, and the frequency of incident dementia was modest. A post hoc analysis of the data from a RCT of another GLP1 analogue, dulaglutide, for the prevention of adverse cardiovascular outcomes in people with diabetes, also showed a significant reduction in incident dementia in participants treated with dulaglutide compared with participants receiving placebo62. The findings of these RCTs need to be interpreted cautiously as they are based on post hoc analyses, but are consistent with a role for GLP1 analogue treatment in preventing the development of dementia.
Several more recent studies of GLP1 analogues in individuals with AD are underway or have been completed. A preliminary randomized, placebo-controlled clinical 18F-fluorodeoxyglucose (FDG)-PET study in 38 individuals with AD demonstrated that, compared with placebo, 6 months of treatment with liraglutide at a dose of 1.8 mg per day by subcutaneous injection prevented a decline in glucose metabolism in the brain63. Glucose metabolism is used as a marker of brain activity, and a lack of decline in glucose metabolism is usually taken to indicate preservation of biological brain function. Further analysis indicated that the underlying mechanism for this effect was an increase in blood–brain glucose transfer capacity and that, in the group of participants who received liraglutide, transfer capacity was the same as in healthy controls. A larger phase II RCT involving 204 participants with AD was completed in 2019 (REF.64). The results of an 18-month pilot double-blind placebo-controlled RCT of exenatide have been reported65. The study, which included only 21 participants, found that exenatide was well-tolerated, although an expected increase in nausea and decreased appetite was observed in the group that received the drug compared with the group that received placebo. The study found no significant difference in clinical, cognitive, neuroimaging or cerebrospinal fluid (CSF) measures between the two groups; however, given the very limited power of this study, these observations cannot be meaningfully interpreted. The levels of Aβ42 in plasma extracellular neuronal vesicles were lower in participants receiving exenatide than in participants receiving placebo, which is an interesting result65.
The results of these studies of GLP1 analogues are promising and provide increasing evidence that these drugs might prevent incident dementia in people with diabetes. A broader question is the potential utility of GLP1 analogues for the treatment of MCI due to AD or AD outside the context of diabetes. The preclinical studies in this area are encouraging, but further trials are needed and the results of the ongoing Evaluating Liraglutide in Alzheimer’s Disease (ELAD) trial are eagerly awaited.
New priority compounds
During the 2018–2019 Delphi process a total of five compounds (or classes of compounds) were nominated for further consideration by at least three members of the panel. These compounds were ACE inhibitors, antiviral drugs, disease-modifying antirheumatic drugs (DMARDs), fasudil and phenserine (TABLE 1). Following several rounds of prioritization, the panel came to a clear consensus that the three highest priority candidates for repurposing in AD were fasudil, antiviral drugs and phenserine. Each of these identified candidates achieved the same prioritization rating, and there was no specific prioritization among the three candidates.
Table 1 |.
Drug classes | Proposed candidates | Proposed mechanism of action | Summary of evidence | Remaining work required |
---|---|---|---|---|
Shortlisted candidates | ||||
ROCK inhibitors | Fasudil | Reduction in Aβ levels in vitro through the Dkk1-driven Wnt–PCP pathway72; reduction in inflammation70; prevention of synaptic damage71 and impaired dendritic arborization69 | Strong and consistent evidence of synaptic protection, reduction in amyloid and cognitive benefits in a range of in vivo animal models of AD67,71,72; several studies have shown acceptable safety in people with pulmonary hypertension and ischaemic heart disease74,136,137; only one very small study in MCI and AD, which found better scores on the verbal fluency test, Mini-Mental State Examination and activities of daily living with fasudil treatment than with nimodipine; the full study has not been published in English75 | A well-powered RCT among participants with AD or MCI is needed to evaluate the effect of fasudil on cognitive function |
AChE inhibitors | Phenserine | Suppression of IL-1β production; reduction in glutamate-induced excitotoxicity; protection against oxidative stress; reduction in Aβ levels; increase in production of BDNF; inhibition of APP and α-synuclein synthesis; and anti-apoptosis action on pre-programmed cell death pathway19 | Several preclinical studies showed that phenserine reduces APP levels in cultured cells and in the brain of animal models83–87; phase II studies of phenserine showed good tolerability and demonstrated some indication of cognitive benefit, although the study was under-powered to properly examine cognitive function78 | Further studies are needed to verify the potential mechanism of action in humans; these studies need to have adequate power to measure cognitive benefits |
Antiviral drugs | Acidovir, pencidovir, valaciclovir and foscarnet | In vitro evidence suggests that HSV can accelerate the accumulation of amyloid97–99 and promote abnormal tau phosphorylation100–102; antiviral drugs might mitigate these effects | A post-mortem case–control study in carriers of APOE ε4 found that AD was more common among individuals who had HSV than in those who did not95; an epidemiological study also showed that a cohort of persons with HSV had a higher risk of developing dementia than those without HSV104; recent large-scale studies suggest that the association between HSV and dementia is mitigated or reversed by antiviral therapy94,104 | At least two small RCTs in a combined total of 163 individuals with AD are in progress138,139, but a well-powered RCT is needed |
Non-shortlisted compounds | ||||
DMARDs | Methotrexate, chloroquine phosphate, proguanil hydrochloride, cyclosporine, cyclophosphamide, hydroxychloroquine sulfate and sodium aurothiomalate | The potent anti-inflammatory actions of this class of agents might be a potential mechanism of action, but this has not been clarified in preclinical studies | A population-based retrospective cohort study found that participants using DMARDs had a modestly reduced risk of dementia compared with participants not using DMARDs18; a double-blind RCT in 168 individuals with mild AD over 18 months showed that hydroxychloroquine did not prevent cognitive decline107 compared with placebo; an open-label trial in ten individuals with AD treated with hydroxychloroquine showed that CSF levels of Aβ did not change after treatment140 (reviewed elsewhere107) | More robust preclinical studies are needed to establish mechanism of action; high-powered RCTs are also needed to confirm findings from observational studies |
ACE inhibitors | Captopril, ramipril, lisinopril and perindopril | Reduction in amyloid deposition and tau hyperphosphrylation141; protection against oxidative stress109,142; reduction in blood pressure | Evidence of benefit inconsistent across studies143 | Although there is some supportive preclinical evidence, the epidemiological evidence is fairly weak; RCTs, several of which are already ongoing, are needed to distinguish between the effect of hypertension control and the specific effects of ACE inhibitors |
Aβ, amyloid-β; ACE, angiotensin-converting enzyme; AChE, acetylcholinesterase; AD, Alzheimer disease; APP, amyloid precursor protein; BDNF, brain-derived neurotrophic factor; CSF, cerebrospinal fluid; DMARDs, disease-modifying antirheumatic drugs; HSV, herpes simplex virus; MCI, mild cognitive impairment; RCT, randomized clinical trial; ROCK, rho kinase.
Fasudil
Fasudil, a selective inhibitor of rho kinase (ROCK) 1 and 2, is a potent vasodilator, particularly of the cerebral vasculature66, and is approved in Japan and China for the treatment of cerebral vasospasm following subarachnoid haemorrhage67. Fasudil was first suggested as a potential treatment for AD in 2009 when a study found that administration of the compound was associated with protection against age-related memory impairment in rats68. In a subsequent study, fasudil was mixed into artificial CSF administered directly into the brain in the APP–PS1 mouse model of AD. The aberrant dendritic arborization phenotype of this mouse model was reduced in mice receiving fasudil compared with mice receiving artificial CSF alone69. Fasudil administration was also associated with protection against hippocampal neurodegeneration induced by intracerebroventricular injection of Aβ1–42 in rats. The authors reported increased IL‐1β, increased tumour necrosis factor (TNF) production and increased activation of NF‐κB in rats receiving fasudil treatment compared with rats receiving placebo and postulated that the protection against amyloid might be related to suppression of inflammatory responses70. More recent work using cell culture and several different transgenic mouse models of AD suggests that fasudil can protect against synaptic loss and cognitive impairment mediated by Aβ through the Dkk1-driven Wnt–PCP pathway67,71. Fasudil, delivered intraperitoneally, was also associated with reduced brain amyloid burden in the 3×AD-TG mouse model of AD72.
Killick et al. identified 14 randomized placebo-controlled trials of fasudil in the literature73. These trials included a combined total of >500 participants with a range of indications from coronary heart disease to pulmonary hypertension. Fasudil was administered at doses of 60–240 mg per day, and most trials found good tolerability with no significant safety concerns. However, one double-blind, placebo-controlled clinical trial of a new extended release formulation of fasudil for pulmonary arterial hypertension did highlight several safety concerns74. In this trial, of 12 patients in the active treatment group, treatment was discontinued in one because of renal impairment and one patient died from heart failure. One small 2-month RCT conducted in China investigated the efficacy of fasudil for the treatment of AD75. In this trial, 106 male participants with MCI treated with nimodopine were randomly assigned to receive either 30 mg intravenous fasudil (once per day) or placebo for 2 months. Preliminary results indicated that fasudil was well tolerated, and the group treated with fasudil had significantly higher MMSE scores than the group that received placebo. This efficacy data should be interpreted cautiously, but good tolerability in individuals with MCI is important.
Overall, there is high concordance in the results of different preclinical studies, which suggest that fasudil targets classic AD neuropathology76 by reducing amyloid burden and also targets other pathological mechanisms that contribute to AD, for example, by protecting against inflammation and synaptic damage76,77. These biochemical and physiological benefits have consistently translated into cognitive improvement in in vivo AD models70,76,77.
Phenserine
Phenserine was initially developed and evaluated as a cholinesterase inhibitor78. However, several mechanisms exist by which phenserine might reduce neuronal and synaptic loss79. which are important pathways in AD, traumatic brain injury and other neurodegenerative diseases. The results of a range of preclinical studies indicate that phenserine suppresses production of IL-1β, reduces glutamate-induced excitotoxicity, protects against H2O2-induced oxidative toxicity, reduces levels of Aβ, improves neural precursor cell viability, elevates brain-derived neurotrophic factor and inhibits APP and α-synuclein synthesis79–82. In particular, the results of several preclinical studies indicate that phenserine can reduce APP levels in vitro and in vivo83–87. Although these potential actions are of interest, more importantly, recent work has suggested that phenserine might confer significant neuroprotection by inhibiting apoptosis via actions on a pre-programmed cell death pathway82. This hypothesis has been evaluated in several rodent models of neuronal loss, including the APP–PSEN1 mouse model of AD, a rat model of post-stroke reperfusion injury and a weight drop mouse model of traumatic brain injury80–82. In all of these animal studies, treatment with phenserine was associated with significant reductions in the severity of neurodegenerative lesions and decreases in the neuroinflammatory response (via suppression of the IBA1 and TNF pathways) in the hippocampus and/or cortex79,81,82. Phenserine treatment was also associated with protection against reductions in synaptic density and levels of synaptophysin in animal models of AD and traumatic brain injury79–82. The multifaceted pharmacological action of phenserine as a neuroprotective agent was an important factor in the prioritization of this compound by the panel. In addition, administration of phenserine was associated with improved cognition in rats with NMDA receptor antagonist-induced impairments in learning88.
Phenserine has been evaluated in two phase II placebo-controlled trials in individuals with mild to moderate AD78,89. The results of a phase II, 12-week RCT in 164 participants with AD indicated that (−)-phenserine (10–15 mg twice per day) had a favourable safety profile and the group of participants receiving the drug showed significantly better cognitive function than the group of participants receiving placebo78. A trend towards improvement in global outcome was observed in participants who received the higher dose of phenserine78,90, with Cohen’s D effect sizes of 0.3–0.4 for symptomatic benefits, which is similar to the effect sizes seen with other cholinesterase inhibitors91. A second, smaller RCT randomized 20 participants with mild AD to receive either phenserine (15 mg twice per day) or placebo for 3 months89. Over the subsequent 3 months, the patients allocated to phenserine continued to receive phenserine treatment while the placebo group then received donepezil in an open design. At the end of the first 3 months, the group of participants receiving phenserine had significantly better cognitive function (measured with a composite neuropsychological test) than the group of participants receiving placebo, and this significant difference between the two groups was maintained after the group receiving placebo had switched to donepezil for 3 months89. Although these results are encouraging, they must be interpreted cautiously given the small sample size of the study. Furthermore, a phase III trial of phenserine was discontinued early for commercial reasons and did not demonstrate a significant beneficial effect on the primary outcome measures, which were ADAS-Cog score and clinician’s interview-based impression of change with care-giver input (CIBIC+)92. The results of this phase III trial have not been published in full, but a press release described non-significant trends towards improvement with the doses of 10 mg and 15 mg92. These results are difficult to interpret on the basis of the preliminary reports, especially as the study was significantly under-powered to detect changes in cognitive and functional outcomes, with only 284 participants randomized in a 2:2:1 design. In addition, the dosing regimen was probably sub-therapeutic as the compound has a half-life of 5–6 hours, but was only administered twice per day, which led to criticism of the trial design93.
Overall, the preclinical evidence that phenserine has biological effects that are relevant to the treatment of AD and other neurodegenerative conditions is strong. These effects include a newly identified influence on apoptosis. Phenserine also has a good clinical safety profile. Although the results from phase II studies are encouraging, they need to be interpreted cautiously given the small sample sizes and short trial durations. Trials of at least 12 months would be needed to identify disease-modifying effects. The potential of phenserine to combine the symptomatic benefits of a cholinesterase inhibitor with additional disease-modifying actions is, however, an exciting prospect.
Antiviral drugs
The potential role of herpes simplex virus (HSV) as a risk factor or mediating factor in the development of AD emerged as a hypothesis in 1991, when HSV-1 was found in an active form in the brain of a large number of older people94. In 1991, a case–control post-mortem study found an association between HSV-1 infection and an increased risk of AD95. Little progress was made until the 2000s and 2010s, when further studies identified HSV-1 DNA within amyloid plaques in individuals with AD96, and provided evidence for a role of HSV-1 in promoting the accumulation of Aβ97–99 and the abnormal phosphorylation of tau100–102. In 2011, the authors of one study used quantitative immunocytochemistry in a kidney cell in vitro model to demonstrate that the changes in Aβ and phospho-tau production did not occur on initial entry of the virus into the cell, but were related to subsequent viral replication103. In vitro, the antiviral drugs aciclovir (the active form of the prodrug valaciclovir), penciclovir (the active form of the prodrug famciclovir) and foscarnet were associated with reductions in Aβ and phospho-tau accumulation, as well as levels of HSV-1. However, foscarnet had a more modest effect than the other two drugs. The accumulation of phospho-tau was dependent on HSV-1 DNA replication, whereas the accumulation of Aβ was not. This work is important in highlighting mechanisms that could link HSV-1 to the development of AD pathologies and in identifying candidate therapies.
More recently, the results of several epidemiological studies have supported the potential value of antiviral therapies in the treatment of AD. The authors of one study used Taiwan’s National Health Insurance Research Database (NHIRD) to evaluate the records of 33,448 individuals and identified 8,362 individuals with a newly diagnosed HSV infection as well as 25,086 randomly selected sex-matched and age-matched controls without HSV infection104. The adjusted hazard ratio for the development of dementia in the participants with HSV-1 relative to the control participants was 2.6 (P < 0.001). Participants with HSV-1 who were treated with anti-herpetic medication had a significantly lower risk of developing dementia than participants with HSV-1 who were not treated with this medication. The risk of dementia was lower among participants who used anti-herpetic medication for ≥30 days than in participants who used this medication for a shorter duration. Using the same database, a larger study of the records of 78,410 individuals identified a significant but more modest increase in the risk of dementia in participants with varicella zoster virus infection than in participants without the infection. This study also found that treatment with antiviral therapy significantly reduced the risk of developing dementia following the diagnosis of herpes zoster105.
Overall, the evidence from in vitro and post-mortem studies suggests that HSV infection and possibly varicella zoster virus infection are risk factors for AD. Although the absence of substantive in vivo studies is a concern, emerging evidence from large-scale epidemiological studies confirms the association between risk of cognitive decline and HSV or varicella zoster virus infections. The results of these epidemiological studies also suggest that this risk can be mitigated by antiviral therapy. Therefore, strong arguments exist for exploring the potential benefit of antiviral drugs in individuals with AD. An ongoing phase II study of valaciclovir aims to recruit 130 participants with mild AD106. The existing evidence suggests that antiviral compounds might be more effective at diminishing the risk of AD or delaying the onset of AD in people with MCI, than as a treatment for individuals who have already developed AD.
In summary, three main classes of compound have emerged from the Delphi consensus process in 2018–2019: fasudil, phenserine and antiviral drugs. GLP analogues were prioritized by the 2012 Delphi consensus process and remain a high priority candidate for repurposing. The prioritization of these compounds is supported by strong packages of preclinical data, most of which include evidence from a number of different preclinical models. The preclinical data also suggest that each of these compounds can have an effect on multiple AD-related therapeutic targets in addition to amyloid. One advantage of repurposed compounds as opposed to newly developed therapeutic agents is that additional data can be gained from epidemiological studies, clinical cohort studies and clinical trials designed to measure a different outcome. For GLP analogues and antiviral drugs, clinical information from epidemiological studies or clinical trials with different primary outcomes support the potential utility of these drugs for the treatment of AD. However, information from clinical trials of any of the prioritized compounds in individuals with MCI or AD is much more limited. As discussed earlier, several clinical trials of phenserine have been performed, and two phase II trials suggested that in individuals with AD phenserine treatment is associated with improved cognition. However, these results are difficult to interpret because the studies used a suboptimal dose of the compound, were of short duration and had limited statistical power. Almost 500 participants per group are needed to provide reasonable power to detect changes in standard neuropsychology measures in an RCT in individuals with mild to moderate AD11. For GLP analogues, only very small preliminary studies have been performed, although the results of these studies are encouraging. The only reported study of fasudil in individuals with MCI or AD showed good tolerability of the compound, but was too small to allow conclusions to be drawn about the effect of the treatment on cognition. No RCTs of antiviral drugs in individuals with MCI or AD were identified in our literature searches. Therefore, the prioritization of these candidates was predominantly based on the preclinical evidence, but with support from clinical information for most of the compounds.
Compounds not short-listed
Disease-modifying agents for rheumatoid arthritis.
Although the anti-inflammatory action of DMARDs could theoretically reduce neuroinflammation in individuals with AD, the preclinical evidence supporting their usefulness was very limited107. The main evidence in favour of DMARDs was from an epidemiological population-based study that found a reduction in dementia risk in individuals receiving DMARDs compared with individuals not receiving DMARDs; however, the reported survival curves showed that the reduction in incidence of new-onset dementia among DMARD users compared with non-DMARD users was very small18. The study did not assess the effect of any single drug within the DMARD class, which is a limitation as these drugs vary widely in terms of pharmacological action, efficacy and tolerability. Furthermore, a placebo-controlled RCT of DMARDs in individuals with AD had negative findings108. On the basis of this evidence, the Delphi consensus panel concluded that DMARDs should not be prioritized as candidates for clinical trials in individuals with AD.
ACE inhibitors.
Some evidence from preclinical studies suggests that ACE inhibitors can protect against AD pathology; for example, in a transgenic mouse model of AD treatment with perindopril was associated with significantly reduced amyloid and tau burdens and levels of oxidative stress109. The clinical evidence in favour of ACE inhibitors was very weak. An open-label study in 113 individuals with AD110 showed no significant benefits of perindopril treatment. A 4-month double-blind, placebo-controlled, pilot RCT of ramipril in 14 individuals with hypertension at risk of AD found that compared with placebo, treatment with ramipril was not associated with an improvement in cognition or a reduction in CSF levels of Aβ1–42111. These poor preliminary clinical results led the panel to conclude that ACE inhibitors are not high-priority agents for repurposing for AD treatment, although the cardiovascular and cerebrovascular benefits of these drugs might indirectly reduce the risk of AD.
Transcriptional approaches
Above, we have prioritized drugs on the basis of their established mechanisms of action. Strategies for identifying novel compounds for preclinical testing and clinical trials include transcriptional profiling, which could also be applied to the identification of drugs for repurposing. Disease or injury can perturb gene expression in a characteristic manner in a specific tissue, creating a ‘transcriptional signature’. If a drug perturbs gene expression in an opposing manner to the disease or injury, it might have therapeutic effects. Therefore, assessing the transcriptional changes induced by libraries of compounds could provide an important way of identifying novel candidates for repurposing. The Broad Institute Connectivity Map (CMAP) collated the transcriptional signatures induced by 1,300 drug-like compounds when applied to three cancer cell lines; importantly the CMAP data reflect responses specific to the known targets of the compounds as well as off-target responses112. The CMAP has been complemented by the LINCS L1000 project, which profiled the changes in 1,000 ‘landmark’ transcripts induced by different compounds and used algorithms to predict the likely changes in expression levels of the non-measured transcripts to generate a full transcriptional signature113. The LINCS L1000 program has generated a database of transcriptional signatures for ~20,000 compounds, ~300 biological agents, and short hairpin RNA and/or cDNA against ~5,000 genes in ~100 human cell lines, including induced pluripotent stem cell (iPSC)-derived cortical neurons. The same approach could be applied to other compound libraries.
Transcriptional profiles are widely available for early, middle and late stages of AD and other dementias114 and for almost all of the interventions, including genetic modifications, that are used to generate animal models of these diseases114,115. However, these data come from a variety of platforms and are hosted in different databases. The searchable, platform-independent expression database (SPIED) was developed to facilitate meta-analysis, with the aim of identifying disease-associated transcriptional perturbations that are common to multiple datasets, including data from post-mortem samples from individuals with AD116,117. This approach has identified shared transcriptional changes within multiple, independent AD-associated transcriptional signatures and the transcriptional signatures associated with other neurodegenerative diseases114. When the AD transcriptional signature was probed in CMAP, 153 drugs that perturb the cancer cell transcriptome in an opposing manner were identified114. Importantly, transcriptional changes that oppose those comprising the AD transcriptional signature were also observed when many of these drugs were applied to human iPSC-derived cortical neurons20. In a further study, transcriptional signatures for early and mild AD were used to probe both the CMAP and LINCS L1000 data, and 78 drugs with a transcriptomic signature that was significantly inversely correlated with the AD transcriptomic signature were identified and screened using six independent in vitro assays that were designed to mimic various aspects of AD pathology118. Of these 78 agents, 19 significantly reduced the AD-associated changes in at least two assays, and eight of these 19 agents were novel candidates known or likely to penetrate the brain. Some interesting candidates identified by this study included the adrenergic α1 receptor antagonist doxazosin, the antibiotic thiostrepton, which is known to have proteasome inhibitor properties, and the histamine H2-receptor antagonist famotidine. In addition to the identification of novel candidates for repositioning, the work supports the hypothesis that transcriptional profiling could be an effective way of identifying or triaging compounds for in vitro screening. For example, other hits included drugs already considered to be repositioning candidates in AD, such as metformin, nabumetone and several flavonoids118.
Future directions
The global transcriptional signatures discussed in the previous section were generated without considering the functions of the individual transcripts or the known mechanisms of drug action. Therefore, this process is a ‘black-box’ approach that operates independently of any mechanism-based hypothesis. Almost 30 risk genes for AD have now been detected119, and the identification of drugs that alter the expression of some of these genes, or the expression of another gene with known therapeutic potential, would enable a hypothesis-driven approach to drug repositioning. There are no well-developed examples of this approach in the AD field, but we briefly discuss three examples from related diseases that highlight the promise of this ‘targeted’ repurposing approach.
Accumulation of glutamate at synapses results in neuronal loss via ‘excitotoxicity’, and this process has been implicated as a causative mechanism in both acute brain injury and chronic neurodegenerative diseases such as AD120. Glutamate accumulation can result from the loss or failure of transporters that recycle this neurotransmitter, and reduced levels of the astrocyte glutamate transporter GLT1 (as known as EAAT2) is a characteristic feature of amyotrophic lateral sclerosis (ALS)121. In a milestone paper, Rothstein et al. postulated that drugs that increase the expression of GLT1 would be neuroprotective in a range of conditions, including ALS122. To test this hypothesis, the authors used neuronal cultures to screen 1,040 FDA-approved drugs and nutritionals and identified agents that increased levels of GLT1. The surprising finding was that the application of β-lactam antibiotics to neuronal cultures at concentrations similar to those in the brains of individuals being treated with these antibiotics increased GLT1 levels via a transcriptional mechanism. Moreover, treatment with the β-lactam ceftriaxone was associated with delayed neuronal loss and increased lifespan in a mouse model of ALS123. Beneficial effects of ceftriaxone have been reported in a wide range of non-clinical studies of pathologies that involve excitotoxicity, including models of AD123. Only one phase III clinical trial has tested the effects of ceftriaxone in neurodegenerative disease. The study cohort consisted of individuals with ALS and no significant differences in survival or functional decline (both primary end points) between the group of participants receiving ceftriaxone and the group of participants receiving placebo were detected124. Nonetheless, these findings are a useful example of a targeted repurposing approach and suggest that a trial of ceftriaxone or a related drug in individuals with AD could have positive results.
As opposed to increasing the expression of a protective gene, other researchers have sought to identify drugs that can reduce the expression of a risk gene. This strategy was recently applied to the search for PD therapies. Reducing α-synuclein transcription might be protective against PD125, and a biological screen of FDA-approved drugs showed that α2-adrenergic agonists, such as salbutamol, suppress α-synuclein transcription126. Moreover, in a preclinical rodent model of PD, salbutamol was associated with some protection against pathology and motor deficits, and analysis of clinical records showed that the risk of developing PD was lower in individuals treated with salbutamol than in individuals not treated with the drug126. This association was confirmed in an independent patient cohort127; however, other researchers have suggested that the association might in part arise from the use of salbutamol to treat smoking-related pulmonary disease, which means that the cohort treated with salbutamol were likely to already have had a reduced risk of developing PD as a result of nicotine exposure128. Future clinical trials are needed to establish the effects of salbutamol on PD, but nonetheless similar approaches could be used to identify compounds that reduce the expression of AD risk genes.
Boosting levels of endogenous growth factors is another potential therapeutic approach that has been poorly explored in AD, but might be feasible, as shown by several studies in the field of PD129–133. Recombinant human fibroblast growth factor 20 (FGF20) can limit neuronal loss in preclinical models of PD129,130; however, delivery and target engagement of growth factors remains a challenge in the clinical setting131. Endogenous FGF20 is enriched in the nigrostriatal pathway132, and a simple in silico interrogation of CMAP identified 50 FDA-approved drugs that increase FGF20 transcript levels in cancer cell lines, 16 of which had transcriptional profiles that suggest they might be beneficial in PD133. Salbutamol and triflusal were in included in these 16 promising candidates and were then tested in vivo. In the 6-hydroxydopamine rat model of PD, treatment with either salbutamol or triflusal was associated with elevated levels of endogenous FGF20 in the nigrostriatal tract and a degree of neuroprotection. Evidence for salbutamol protecting humans against PD was discussed in the previous paragraph. Triflusal is a trifluoromethyl derivative of acetylsalicylic acid that inhibits platelet aggregation and, thereby, reduces the risk of stroke134. The drug also has anti-inflammatory, anti-excitotoxicity and anti-Zn2+ toxicity effects that might limit ischaemic brain damage135.
Limitations of the targeted repurposing approach include the fact that a drug is likely to alter the expression of perhaps hundreds of transcripts. For example, whether salbutamol is neuroprotective because it reduces α-synuclein expression, increases FGF20 expression, acts via a third unknown mechanism, or acts via a combination of multiple mechanisms is not clear. Likewise, triflusal could be neuroprotective in PD because it elevates FGF20 and/or because it has antioxidant and anti-inflammatory properties and/or because it acts via other unknown mechanisms. Similarly, although the parsimonious explanation for the neuroprotective properties of β-lactam antibiotics is an increase in glutamate uptake122, these drugs also have antioxidant and metal-chelating properties that might explain or contribute to their efficacy as neuroprotective drugs123. This targeted repurposing approach is still in its infancy — transcriptional profiles have been successful in predicting some effects of compounds in vitro and in vivo, but it will be several years before we have any proof-of-concept clinical trials or examples of clinically available treatments. Nonetheless, the hypothesis-driven nature of targeted repurposing facilitates the design of experiments to directly test postulated mechanism of action of a specific compound.
Conclusions
Drug repositioning or repurposing offers an attractive and cost-effective approach that can complement traditional drug development. We used a Delphi consensus process to identify promising classes of compound for repurposing that we feel merit evaluation in clinical trials. GLP1 analogues were identified as priority compounds in a Delphi consensus in 2012 (REF.9), but in this Review we discussed further supportive evidence that has subsequently emerged. We also presented and discussed three new compounds or classes of compound that were prioritized by the new Delphi consensus process. These compounds include the ROCK2 inhibitor fasudil, the cholinesterase inhibitor phenserine, which also has novel anti-apoptotic properties, and the antiviral drugs aciclovir, valaciclovir and famciclovir. We also reviewed the evidence for a novel transcriptomic approach to drug repurposing that could substantially increase the scale of identification of candidate compounds.
The potential advantages of complementing traditional drug discovery approaches with drug repositioning or repurposing include reduced costs and faster approval. However, several challenges to the expansion of this field remain, including the need for novel methodologies to identify and screen new candidates, for example, transcriptomic approaches. Creating and expanding funding streams to prioritize this work and providing better commercial incentives for repurposing, perhaps through better protection by use patents, will also be important.
Key points.
Drug repositioning and repurposing offers a valuable alternative route for the identification of effective disease-modifying treatments for Alzheimer disease (AD).
The Delphi method can be used to bring together the opinion of multiple experts to suggest candidates for repurposing.
An expert Delphi consensus published in 2012 prioritized five compounds for repurposing as treatments for AD, of which glucagon-like peptide analogues remain high priority candidates.
A Delphi consensus involving the authors of this Review was conducted in 2018–2019 and identified the ROCK inhibitor fasudil, the cholinesterase inhibitor phenserine and antiviral treatments such as valaciclovir as high priority candidates for trials in individuals with AD.
The prioritization of these compounds was supported by strong packages of preclinical data, most of which include evidence from a number of different preclinical models.
Transcriptional screening approaches offer a novel means of identifying potential treatment candidates by targeting AD-associated transcriptional profiles.
Acknowledgements
The authors thank J. Pickett from the Alzheimer’s Society (UK), C. Routledge from Alzheimer Research UK and H. Fillit from the Alzheimer’s Drug Discovery Foundation for their invaluable contributions to the 2018–2019 Delphi consensus. We also thank the Wellcome Trust for supporting much of the work pertaining to transcript signatures presented in the current paper. J.C. is supported by NIGMS grant P20GM109025, NINDS grant U01NS093334, and NIA grants R01AG053798 and P20AG068053.
Competing interests
C.B. reports grants and personal fees from Acadia Pharmaceutical Company and H. Lundbeck, as well as personal fees from AARP, Biogen, Eli Lilly, Novo Nordisk, Otsuka, Roche and Synexus, outside the submitted work. D.A. has received research support and/or honoraria from Astra-Zeneca, GE Health, H. Lundbeck and Novartis Pharmaceuticals, and served as a paid consultant for Biogen, Eisai, H. Lundbeck, Heptares and Mentis Cura. J.C. reports grants from National Institute of General Medical Sciences Centers of Biomedical Research Excellence (grant no. P20GM109025), during the conduct of the study; personal fees from Acadia, Actinogen, AgeneBio, Alkahest, Alzheimer Drug Discovery Foundation, Alzheon, Avanir, Axsome, Biogen, Cassava, Cerecin, Cerevel, Cognoptix, Cortexyme, EIP Pharma, Eisai, Foresight, Green Valley, Grifols, Hisun, Karuna, Nutricia, Otsuka, reMYND, Resverlogix, Roche, Samumed, Samus Therapeutics, Third Rock, Signant Health, Sunovion, Suven, and United Neuroscience pharmaceutical and assessment companies; other from ADAMAS, BioAsis, MedAvante, QR Pharma, United Neuroscience, other from Keep Memory Alive; outside the submitted work. J.C. has copyright for the Neuropsychiatric Inventory with royalties paid. J.O’B. reports personal fees from Axon, Eisai, GE Healthcare, Roche and TauRx; grants and personal fees from Avid Technology and Eli Lilly; grants from Alliance Medical and Merck, outside the submitted work. J.L.M. reports personal fees from Alector, Alergan, Eisai, Genentech, Oryzon, Green Valley, H. Lundbeck, Janssen, Eli Lilly, MSD, Novartis and Raman Health; personal fees and other from Biogen, outside the submitted work. All other authors declare no competing interests.
Footnotes
Review criteria
Searches were performed in EMBASE, PsycINFO, MEDLINE and Cochrane databases for papers published after 1960. Search terms were as follows: generic class OR specific drug names OR any known alternative name (obtained from the electronic Medicines Compendium and the British National Formulary) AND dement* OR Alzheim* OR mild cognitive impairmen* OR neuropsych* test* OR cognitive func*.
References
- 1.Alzheimer’s Disease International. World Alzheimer Report 2016. https://www.alz.co.uk/research/world-report-2016 (2016).
- 2.Petersen RC Mild cognitive impairment. Continuum 22, 404–418 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Jack CR et al. Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol. 12, 207–216 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Khan A et al. Emerging treatments for Alzheimer’s disease for non-amyloid and non-tau targets. Expert. Rev. Neurother 17, 683–695 (2017). [DOI] [PubMed] [Google Scholar]
- 5.Morris GP, Clark IA & Vissel B Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer’s disease. Acta Neuropathol. Commun 2, 135 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Honig LS et al. Trial of solanezumab for mild dementia due to Alzheimer’s disease. N. Engl. J. Med 378, 321–330 (2018). [DOI] [PubMed] [Google Scholar]
- 7.Egan MF et al. Randomized trial of verubecestat for prodromal Alzheimer’s disease. N. Engl. J. Med 380, 1408–1420 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ballard C & Murphy C Clinical trial pipelines for Alzheimer’s disease pose challenges for future effective treatment and therapies [abstract P4–017]. Alzheimers Dement. 14, P1439 (2018). [Google Scholar]
- 9.Corbett A et al. Drug repositioning for Alzheimer’s disease. Nat. Rev. Drug Discov 11, 833–846 (2012). [DOI] [PubMed] [Google Scholar]
- 10.Hawkes N Pfizer abandons research into Alzheimer’s and Parkinson’s diseases. BMJ 360, k122 (2018). [DOI] [PubMed] [Google Scholar]
- 11.Alzheimer’s Disease Neuroimaging Initiative. More ADNI results 2: it’s all about power AlzForum https://www.alzforum.org/news/research-news/more-adni-results-2-its-all-about-power (2008).
- 12.Biogen. Aducanumab update. Investors.biogen.com https://investors.biogen.com/static-files/5a31a1e3-4fbb-4165-921a-f0ccb1d64b65 (2019). [Google Scholar]
- 13.Loera-Valencia R et al. Current and emerging avenues for Alzheimer’s disease drug targets. J. Intern. Med 286, 398–437 (2019). [DOI] [PubMed] [Google Scholar]
- 14.Langedijk J et al. Drug repositioning and repurposing: terminology and definitions in literature. Drug Discov. Today 20, 1027–1034 (2015). [DOI] [PubMed] [Google Scholar]
- 15.Stewart AK Medicine. How thalidomide works against cancer. Science 343, 256–257 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hubsher G, Haider M & Okun MS Amantadine: the journey from fighting flu to treating Parkinson disease. Neurology 78, 1096–1099 (2012). [DOI] [PubMed] [Google Scholar]
- 17.Scott TJ, O’Connor AC, Link AN & Beaulieu TJ Economic analysis of opportunities to accelerate Alzheimer’s disease research and development. Ann. N. Y. Acad. Sci 1313, 17–34 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Judge A et al. Protective effect of antirheumatic drugs on dementia in rheumatoid arthritis patients. Alzheimers Dement. 3, 612–621 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Seyb KI et al. Identification of small molecule inhibitors of β-amyloid cytotoxicity through a cell-based high-throughput screening platform. J. Biomol. Screen 13, 870–878 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Williams G et al. Drug repurposing for Alzheimer’s disease based on transcriptional profiling of human iPSC-derived cortical neurons. Transl Psychiatry. 9, 220 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Howard R et al. Minocycline at 2 different dosages vs placebo for patients with mild Alzheimer disease: a randomized clinical trial. JAMA Neurol. 77, 164–174 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lawlor B et al. NILVAD Study Group. Nilvadipine in mild to moderate Alzheimer disease: a randomised controlled trial. PLoS Med. 15, e1002660 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kehoe PG et al. Results of the reducing pathology in Alzheimer’s disease through angiotensin targeting (RADAR) trial [abstract LB3]. J. Prev. Alzheimers Dis 6, S31–S32 (2019). [DOI] [PubMed] [Google Scholar]
- 24.McKhann G et al. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology 34, 939–944 (1984). [DOI] [PubMed] [Google Scholar]
- 25.Birks JS Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Databast Syst. Rev 1, CD005593 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Howard R et al. Determining the minimum clinically important differences for outcomes in the DOMINO trial. Int. J. Geriatr. Psychiatry 26, 812–817 (2011). [DOI] [PubMed] [Google Scholar]
- 27.Kume K et al. Effects of telmisartan on cognition and regional cerebral blood flow in hypertensive patients with Alzheimer’s disease. Geriatr. Gerontol. Int 12, 207–214 (2012). [DOI] [PubMed] [Google Scholar]
- 28.Kehoe PG, Miners JS & Love S Angiotensins in Alzheimer’s disease — friend or foe? Trends Neurosci. 32, 619–628 (1997). [DOI] [PubMed] [Google Scholar]
- 29.Kehoe PG & Passmore PA The renin–angiotensin system and antihypertensive drugs in Alzheimer’s disease: current standing of the angiotensin hypothesis? J. Alzheimers Dis 30, S251–S269 (2012). [DOI] [PubMed] [Google Scholar]
- 30.Wright JW & Harding JW Brain renin-angiotensin — a new look at an old system. Prog. Neurobiol 95, 49–67 (2011). [DOI] [PubMed] [Google Scholar]
- 31.Culman J, Blume A, Gohlke P & Unger T The renin-angiotensin system in the brain: possible therapeutic implications for AT1-receptor blockers. J. Hum. Hypertens 16, S64–S70 (2002). [DOI] [PubMed] [Google Scholar]
- 32.Wang J et al. Valsartan lowers brain β-amyloid protein levels and improves spatial learning in a mouse model of Alzheimer disease. J. Clin. Invest 117, 3393–3402 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Mogi M et al. Telmisartan prevented cognitive decline partly due to PPAR-γ activation. Biochem. Biophys. Res. Commun 375, 446–449 (2008). [DOI] [PubMed] [Google Scholar]
- 34.Papadopoulos P, Tong XK, Imboden H & Hamel E Losartan improves cerebrovascular function in a mouse model of Alzheimer’s disease with combined overproduction of amyloid-β and transforming growth factor-β1. J. Cereb. Blood Flow. Metab 37, 1959–1970 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Royea J, Zhang L, Tong XK & Hamel E Angiotensin IV receptors mediate the cognitive and cerebrovascular benefits of losartan in a mouse model of Alzheimer’s disease. J. Neurosci 37, 5562–5573 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ongali B et al. Angiotensin II type 1 receptor blocker losartan prevents and rescues cerebrovascular, neuropathological and cognitive deficits in an Alzheimer’s disease model. Neurobiol. Dis 68, 126–136 (2014). [DOI] [PubMed] [Google Scholar]
- 37.Danielyan L et al. Protective effects of intranasal losartan in the APP/PS1 transgenic mouse model of Alzheimer disease. Rejuvenation Res. 13, 195–201 (2010). [DOI] [PubMed] [Google Scholar]
- 38.Liu X et al. Losartan-induced hypotension leads to tau hyperphosphorylation and memory deficit. J. Alzheimers Dis. 40, 419–427 (2014). [DOI] [PubMed] [Google Scholar]
- 39.Tian M, Zhu D, Xie W & Shi J Central angiotensin II-induced Alzheimer-like tau phosphorylation in normal rat brains. FEBS Lett. 586, 3737–3745 (2012). [DOI] [PubMed] [Google Scholar]
- 40.Zhu D et al. Central angiotensin II stimulation promotes β amyloid production in Sprague Dawley rats. PLoS ONE 6, e16037 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Anderson C et al. Renin-angiotensin system blockade and cognitive function in patients at high risk of cardiovascular disease: analysis of data from the ONTARGET and TRANSCEND studies. Lancet Neurol. 10, 43–53 (2011). [DOI] [PubMed] [Google Scholar]
- 42.Lithell H et al. The study on cognition and prognosis in the elderly (SCOPE): principal results of a randomized double-blind intervention trial. J. Hypertens 21, 875–886 (2003). [DOI] [PubMed] [Google Scholar]
- 43.Livingston G et al. Dementia: prevention, intervention, and care. Lancet 390, 2673–2734 (2017). [DOI] [PubMed] [Google Scholar]
- 44.Williamson JD et al. Effect of intensive vs standard blood pressure control on probable dementia: a randomized clinical trial. JAMA 321, 553–561 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.US National Library of Medicine. ClinicalTrials.gov https://www.clinicaltrials.gov/ct2/show/NCT02471833 (2019). [DOI] [PubMed]
- 46.US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02085265 (2020). [DOI] [PubMed]
- 47.US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02646982 (2020). [DOI] [PubMed]
- 48.Schrijvers EMC et al. Insulin metabolism and the risk of Alzheimer disease — the Rotterdam study. Neurology 75, 1982–1987 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Perry T et al. Glucagon-like peptide-1 decreases endogenous amyloid-β peptide (Aβ) levels and protects hippocampal neurons from death induced by Aβ and iron. J. Neurosci. Res 72, 603–612 (2003). [DOI] [PubMed] [Google Scholar]
- 50.Holscher C in Vitamins and Hormones: Incretins and Insulin Secretion (ed. Litwack G) 331–354 (Academic Press, 2010). [DOI] [PubMed] [Google Scholar]
- 51.Li L et al. (Val8) glucagon-like peptide-1 prevents tau hyperphosphorylation, impairment of spatial learning and ultra-structural cellular damage induced by streptozotocin in rat brains. Eur. J. Pharmacol 674, 280–286 (2012). [DOI] [PubMed] [Google Scholar]
- 52.Perry T et al. A novel neutrotrophic property of glucagon-like peptide 1: a promoter of nerve growth factor-mediated differentiation in PC12 cells. J. Pharmacol. Exp. Ther 300, 958–966 (2002). [DOI] [PubMed] [Google Scholar]
- 53.Wang XH et al. Val8-glucagon-like peptide-1 protects against Aβ1–40-induced impairment of hippocampal late-phase long-term potentiation and spatial learning in rats. Neuroscience 170, 1239–1248 (2010). [DOI] [PubMed] [Google Scholar]
- 54.Kastin AJ, Akerstrom V & Pan W Interactions of glucagon-like peptide-1 (GLP-1) with the blood–brain barrier. J. Mol. Neurosci 18, 7–14 (2002). [DOI] [PubMed] [Google Scholar]
- 55.McClean PL, Parthsarathy V, Faivre E & Holscher C The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer’s disease. J. Neurosci 31, 6587–6594 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Gengler S, McClean PL, McCurtin R, Gault VA & Hölscher C Val(8)GLP-1 rescues synaptic plasticity and reduces dense core plaques in APP/PS1 mice. Neurobiol. Aging 33, 265–276 (2012). [DOI] [PubMed] [Google Scholar]
- 57.Hamilton A, Patterson S, Porter D, Gault VA & Holscher C Novel GLP-1 mimetics developed to treat type 2 diabetes promote progenitor cell proliferation in the brain. J. Neurosci. Res 89, 481–489 (2011). [DOI] [PubMed] [Google Scholar]
- 58.McClean PL, Jalewa J & Hölscher C Prophylactic liraglutide treatment prevents amyloid plaque deposition, chronic inflammation and memory impairment in APP/PS1 mice. Behav. Brain Res 293, 96–106 (2015). [DOI] [PubMed] [Google Scholar]
- 59.Qi L et al. Subcutaneous administration of liraglutide ameliorates learning and memory impairment by modulating tau hyperphosphorylation via the glycogen synthase kinase-3β pathway in an amyloid β protein induced Alzheimer disease mouse model. Eur. J. Pharmacol 783, 23–32 (2016). [DOI] [PubMed] [Google Scholar]
- 60.Hansen HH et al. The GLP-1 receptor agonist liraglutide improves memory function and increases hippocampal CA1 neuronal numbers in a senescence-accelerated mouse model of Alzheimer’s disease. J. Alzheimers Dis 46, 877–888 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Xiong H et al. The neuroprotection of liraglutide on Alzheimer-like learning and memory impairment by modulating the hyperphosphorylation of tau and neurofilament proteins and insulin signaling pathways in mice. J. Alzheimers Dis 37, 623–635 (2013). [DOI] [PubMed] [Google Scholar]
- 62.Cukierman-Yaffe T et al. Effect of dulaglutide on cognitive impairment in type 2 diabetes: an exploratory analysis of the REWIND trial. Lancet Neurol. 19, 582–590 (2020). [DOI] [PubMed] [Google Scholar]
- 63.Gejl M et al. Blood-brain glucose transfer in Alzheimer’s disease: effect of GLP-1 analog treatment. Sci. Rep 7, 17490 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01843075 (2019). [DOI] [PubMed]
- 65.Mullins RJ et al. A pilot study of exenatide actions in Alzheimer’s disease. Curr. Alzheimer Res 16, 741–752 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ono-Saito N, Niki I & Hidaka H H-series protein kinase inhibitors and potential clinical applications. Pharmacol. Ther 82, 123–131 (1999). [DOI] [PubMed] [Google Scholar]
- 67.Sellers KJ et al. Amyloid β synaptotoxicity is Wnt-PCP dependent and blocked by fasudil. Alzheimers Dement 14, 306–317 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Huentelman MJ et al. Peripheral delivery of a ROCK inhibitor improves learning and working memory. Behav. Neurosci 123, 218–223 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Couch BA, DeMarco GJ, Gourley SL & Koleske AJ Increased dendrite branching in AβPP/PS1 mice and elongation of dendrite arbors by fasudil administration. J. Alzheimers Dis 20, 1003–1008 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Song Y, Chen X, Wang LY, Gao W & Zhu MJ Rho kinase inhibitor fasudil protects against β-amyloid-induced hippocampal neurodegeneration in rats. CNS Neurosci. Ther 19, 603–610 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Killick R et al. Clusterin regulates β-amyloid toxicity via Dickkopf-1-driven induction of the wnt-PCP-JNK pathway. Mol. Psychiatry 19, 88–98 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Elliott C et al. A role for APP in Wnt signalling links synapse loss with β-amyloid production. Transl Psychiatry 8, 179 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Killick R, Ballard C, Aarsland D & Sultana J Fasudil for the treatment of Alzheimer’s disease. Preprint at OSF https://osf.io/3e4y6 (2020).
- 74.Fukumoto Y et al. Double-blind, placebo-controlled clinical trial with a rho-kinase inhibitor in pulmonary arterial hypertension. Circ. J 77, 2619–2625 (2013). [DOI] [PubMed] [Google Scholar]
- 75.Yan B et al. Curative effect of fasudil injection combined with nimodipine on Alzheimer disease of elderly patients. J. Clin. Med. Pract 14, 36 (2011). [Google Scholar]
- 76.Yu JZ et al. Multitarget therapeutic effect of fasudil in APP/PS1 transgenic mice. CNS Neurol. Disord. Drug Targets 16, 199–209 (2017). [DOI] [PubMed] [Google Scholar]
- 77.Hou Y et al. Changes in hippocampal synapses and learning-memory abilities in a streptozotocin-treated rat model and intervention by using fasudil hydrochloride. Neuroscience 200, 120–129 (2012). [DOI] [PubMed] [Google Scholar]
- 78.Winblad B et al. Phenserine efficacy in Alzheimer’s disease. J. Alzheimers Dis 22, 1201–1208 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Becker RE et al. Phenserine and inhibiting pre-programmed cell death: in pursuit of a novel intervention for Alzheimer’s disease. Curr. Alzheimer Res 15, 883–891 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Chang C-F et al. (−)-Phenserine inhibits neuronal apoptosis following ischemia/reperfusion injury. Brain Res. 1677, 118–128 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Tweedie D et al. Cognitive impairments induced by concussive mild traumatic brain injury in mouse are ameliorated by treatment with phenserine via multiple non-cholinergic and cholinergic mechanisms. PLoS ONE 11, e0156493 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Lecca D et al. (−)-Phenserine and the prevention of pre-programmed cell death and neuroinflammation in mild traumatic brain injury and Alzheimer’s disease challenged mice. Neurobiol. Dis 130, 104528 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Sugaya K et al. Practical issues in stem cell therapy for Alzheimer’s disease. Curr. Alzheimer Res 4, 370–377 (2007). [DOI] [PubMed] [Google Scholar]
- 84.Marutle A et al. Modulation of human neural stem cell differentiation in Alzheimer (APP23) transgenic mice by phenserine. Proc. Natl Acad. Sci. USA 104, 12506–12511 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Lahiri DK et al. Differential effects of two hexahydropyrroloindole carbamate-based anticholinesterase drugs on the amyloid beta protein pathway involved in Alzheimer’s disease. Neuromolecular Med. 9, 157–168 (2007). [DOI] [PubMed] [Google Scholar]
- 86.Lahiri DK et al. The experimental Alzheimer’s disease drug posiphen [(+)-phenserine] lowers amyloid-β peptide levels in cell culture and mice. J. Pharmacol. Exp. Ther 320, 386–396 (2007). [DOI] [PubMed] [Google Scholar]
- 87.Haroutunian V et al. Pharmacological modulation of Alzheimer’s β-amyloid precursor protein levels in the CSF of rats with forebrain cholinergic system lesions. Brain Res. Mol. Brain Res 46, 161–168 (1997). [DOI] [PubMed] [Google Scholar]
- 88.Patel N et al. Phenserine, a novel acetylcholinesterase inhibitor, attenuates impaired learning of rats in a 14-unit T-maze induced by blockade of the N-methyl-D-aspartate receptor. Neuroreport 9, 171–176 (1998). [DOI] [PubMed] [Google Scholar]
- 89.Kadir A et al. Effect of phenserine treatment on brain functional activity and amyloid in Alzheimer’s disease. Ann. Neurol 63, 621–631 (2008). [DOI] [PubMed] [Google Scholar]
- 90.Greig NH et al. An overview of phenserine tartrate, a novel acetylcholinesterase inhibitor for the treatment of Alzheimer’s disease. Curr. Alzheimer Res 2, 281–290 (2005). [DOI] [PubMed] [Google Scholar]
- 91.Birks JS & Harvey RJ Donepezil for dementia due to Alzheimer’s disease. Cochrane Database Syst. Rev 6, CD001190 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Axonyx. Axonyx announces results of curtailed phase III clinical trials for phenserine in Alzheimer’s disease. Businesswire https://www.businesswire.com/news/home/20050920005633/en/Axonyx-Announces-Results-Curtailed-Phase-III-Clinical (2005).
- 93.Schneider LS & Lahiri DK The perils of Alzheimer’s drug development. Curr. Alzheimer Res 6, 77–78 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Jamieson GA, Maitland NJ, Wilcock GK, Craske J & Itzhaki RF Latent herpes simplex virus type 1 in normal and Alzheimer’s disease brains. J. Med. Virol 33, 224–227 (1991). [DOI] [PubMed] [Google Scholar]
- 95.IItzhaki RF et al. Herpes simplex virus type 1 in brain and risk of Alzheimer’s disease. Lancet 349, 241–244 (1997). [DOI] [PubMed] [Google Scholar]
- 96.Wozniak MA, Frost AL & Itzhaki RF Alzheimer’s disease-specific tau phosphorylation is induced by herpes simplex virus type 1. J. Alzheimers Dis 16, 341–350 (2009). [DOI] [PubMed] [Google Scholar]
- 97.Santana S, Recuero M, Bullido MJ, Valdivieso F & Aldudo J Herpes simplex virus type I induces the accumulation of intracellular β-amyloid in autophagic compartments and the inhibition of the nonamyloidogenic pathway in human neuroblastoma cells. Neurobiol. Aging 33, 19–33 (2012). [DOI] [PubMed] [Google Scholar]
- 98.Wozniak MA, Itzhaki RF, Shipley SJ & Dobson CB Herpes simplex virus infection causes cellular β-amyloid accumulation and secretase upregulation. Neurosci. Lett 429, 95–100 (2007). [DOI] [PubMed] [Google Scholar]
- 99.Piacentini R et al. HSV-1 promotes Ca2+-mediated APP phosphorylation and Aβ accumulation in rat cortical neurons. Neurobiol. Aging. 32, 2323.e13–2323.e26 (2011). [DOI] [PubMed] [Google Scholar]
- 100.Wozniak MA, Mee AP & Itzhaki RF Herpes simplex virus type 1 DNA is located within Alzheimer’s disease amyloid plaques. J. Pathol 217, 131–138 (2009). [DOI] [PubMed] [Google Scholar]
- 101.Lerchundi R et al. Tau cleavage at D421 by caspase-3 is induced in neurons and astrocytes infected with herpes simplex virus type 1. J. Alzheimers Dis 23, 513–520 (2011). [DOI] [PubMed] [Google Scholar]
- 102.Zambrano A et al. Neuronal cytoskeletal dynamic modification and neurodegeneration induced by infection with herpes simplex virus type 1. J. Alzheimers Dis 14, 259–269 (2008). [DOI] [PubMed] [Google Scholar]
- 103.Wozniak MA, Frost AL, Preston CM & Itzhaki RF Antivirals reduce the formation of key Alzheimer’s disease molecules in cell cultures acutely infected with herpes simplex virus type 1. PLoS ONE 6, e25152 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Tzeng NS et al. Anti-herpetic medications and reduced risk of dementia in patients with herpes simplex virus infections–a nationwide, population-based cohort study in Taiwan. Neurotherapeutics 15, 417–429 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Chen VC et al. Herpes zoster and dementia: a nationwide population-based cohort study. J. Clin. Psychiatry 79, 16m11312 (2018). [DOI] [PubMed] [Google Scholar]
- 106.Devanand DP et al. Antiviral therapy: valacyclovir treatment of Alzheimer’s disease (VALAD) trial: protocol for a randomised, double-blind, placebo-controlled, treatment trial. BMJ Open 10, e032112 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Sultana J & Ballard C Disease-modifying agents for rheumatoid arthritis: methotrexate, chloroquine phosphate/proguanil hydrochloride, cyclosporine, cyclophosphamide, hydroxychloroquine sulphate, sodium aurothiomalate, and sulfasalazine as potential repurposing treatments for Alzheimer’s Disease; a comprehensive literature review. Preprint at OSF https://mfr.osf.io/render?url=https%3A%2F%2Fosf.io%2F6g57a%2Fdownload (2020). [Google Scholar]
- 108.Van Gool WA et al. Effect of hydroxychloroquine on progression of dementia in early Alzheimer’s disease: an 18-month randomised, double-blind, placebo-controlled study. Lancet 358, 455–460 (2001). [DOI] [PubMed] [Google Scholar]
- 109.Ali MR et al. Tempol and perindopril protect against lipopolysaccharide-induced cognition impairment and amyloidogenesis by modulating brain-derived neurotropic factor, neuroinflammation and oxidonitrosative stress. Naunyn Schmiedebergs Arch. Pharmacol 389, 637–656 (2016). [DOI] [PubMed] [Google Scholar]
- 110.de Oliveira F et al. Brain penetrating angiotensin converting enzyme inhibitors and cognitive change in patients with dementia due to Alzheimer’s disease. J. Alzheimers Dis 42, S321–S324 (2014). [DOI] [PubMed] [Google Scholar]
- 111.Wharton W et al. The effects of ramipril in individuals at risk for Alzheimer’s disease: results of a pilot clinical trial. J. Alzheimers Dis 32, 147–156 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Lamb J et al. The connectivity map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313, 1929–1935 (2006). [DOI] [PubMed] [Google Scholar]
- 113.Subramanian A et al. A next generation connectivity map: L1000 platform and the first 1,000,000 profiles. Cell. 171, 1437–1452.e17 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Gatt A et al. Alzheimer’s disease progression in the 5×FAD mouse captured with a multiplex gene expression array. J. Alzheimer’s Dis 72, 1177–1191 (2019). [DOI] [PubMed] [Google Scholar]
- 115.Hargis KE & Blalock EM Transcriptional signatures of brain aging and Alzheimer’s disease: what are our rodent models telling us? Behav. Brain Res 322, 311–328 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Williams G A searchable cross-platform gene expression database reveals connections between drug treatments and disease. BMC Genomics 13, 12 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Williams G SPIEDw: a searchable platform-independent expression database web tool. BMC Genomics 14, 765 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Ballard C et al. Identifying novel candidates for re-purposing as potential therapeutic agents for AD. Preprint at bioRxiv https://www.biorxiv.org/content/10.1101/622308v1 (2019).
- 119.Bertram L & Tanzi RE Alzheimer disease risk genes: 29 and counting. Nat. Rev. Neurol 15, 191–192 (2019). [DOI] [PubMed] [Google Scholar]
- 120.Maragakis NJ & Rothstein JD Glutamate transporters in neurologic disease. Arch. Neurol 58, 365–370 (2001). [DOI] [PubMed] [Google Scholar]
- 121.Rothstein JD et al. Selective loss of glial glutamate transporter GLT-1 in amyotrophic lateral sclerosis. Ann. Neurol 38, 73–84 (1995). [DOI] [PubMed] [Google Scholar]
- 122.Rothstein JD et al. β-Lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature 433, 73–77 (2005). [DOI] [PubMed] [Google Scholar]
- 123.Yimer EM, Hishe HZ & Tuem KB Repurposing of the β-lactam antibiotic, ceftriaxone for neurological disorders: a review. Front. Neurosci 13, 236 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Cudkowicz ME et al. Safety and efficacy of ceftriaxone for amyotrophic lateral sclerosis: a multi-stage, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 13, 1083–1091 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Singleton AB et al. α-Synuclein locus triplication causes Parkinson’s disease. Science 302, 841 (2003). [DOI] [PubMed] [Google Scholar]
- 126.Mittal S et al. β2-Adrenoreceptor is a regulator of the α-synuclein gene driving risk of Parkinson’s disease. Science 357, 891–898 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Gronich N et al. β2-adrenoceptor agonists and antagonists and risk of Parkinson’s disease. Mov. Disord 33, 1465–1471 (2018). [DOI] [PubMed] [Google Scholar]
- 128.Searles Nielsen S et al. β2-adrenoreceptor medications and risk of Parkinson disease. Ann. Neurol 84, 683–693 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Boshoff EL, Fletcher EJR & Duty S Fibroblast growth factor 20 is protective towards dopaminergic neurons in vivo in a paracrine manner. Neuropharmacology 137, 156–163 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Niu J et al. Efficient treatment of Parkinson’s disease using ultrasonography-guided rhFGF20 proteoliposomes. Drug Deliv. 25, 1560–1569 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Weissmiller AM & Wu C Current advances in using neurotrophic factors to treat neurodegenerative disorders. Transl Neurodegener. 1, 14 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Ohmachi S et al. FGF-20, a novel neurotrophic factor, preferentially expressed in the substantia nigra pars compacta of rat brain. Biochem. Biophys. Res. Commun 277, 355–360 (2000). [DOI] [PubMed] [Google Scholar]
- 133.Fletcher EJR et al. Targeted repositioning identifies drugs that increase fibroblast growth factor 20 production and protect against 6-hydroxydopamineinduced nigral cell loss in rats. Sci. Rep 9, 8336 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Murdoch D & Plosker GL Triflusal: a review of its use in cerebral infarction and myocardial infarction, and as thromboprophylaxis in atrial fibrillation. Drugs 66, 671–692 (2006). [DOI] [PubMed] [Google Scholar]
- 135.Kim SW et al. Neuroprotective effect of triflusal and its main metabolite, 2-hydroxy-4-trifluoromethylbenzoic acid (HTB), in the postischemic brain. Neurosci. Lett 643, 59–64 (2017). [DOI] [PubMed] [Google Scholar]
- 136.Vicari RM et al. Efficacy and safety of fasudil in patients with stable angina: a double-blind, placebo-controlled, phase 2 trial. J. Am. Coll. Cardiol 46, 1803–1811 (2005). [DOI] [PubMed] [Google Scholar]
- 137.Kamei S, Oishi M & Takasu T Evaluation of fasudil hydrochloride treatment for wandering symptoms in cerebrovascular dementia with 31P-magnetic resonance spectroscopy and Xe-computed tomography. Clin. Neuropharmacol 19, 428–438 (1996). [DOI] [PubMed] [Google Scholar]
- 138.US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02997982 (2020). [DOI] [PubMed]
- 139.US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03282916 (2020). [DOI] [PubMed]
- 140.Nitsch RM, Deng M, Tennis M, Schoenfeld D & Growdon JH The selective muscarinic M1 agonist AF102B decreases levels of total Abeta in cerebrospinal fluid of patients with Alzheimer’s disease. Ann. Neurol 48, 913–918 (2000). [PubMed] [Google Scholar]
- 141.AbdAlla S, El Hakim A, Abdelbaset A, Elfaramawy Y & Quitterer U Inhibition of ACE retards tau hyperphosphorylation and signs of neuronal degeneration in aged rats subjected to chronic mild stress. Biomed. Res. Int 2015, 917156 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.AbdAlla S, Langer A, Fu X & Quitterer U ACE inhibition with captopril retards the development of signs of neurodegeneration in an animal model of Alzheimer’s disease. Int. J. Mol. Sci 14, 16917–16942 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Sultana J & Ballard C ACE inhibitors: captopril, ramipril, lisinopril, perindopril as potential repurposing treatments for Alzheimer’s disease; a comprehensive literature review. Preprint at OSF https://mfr.osf.io/render?url=https%3A%2F%2Fosf.io%2Fz9uc4%2Fdownload (2020). [Google Scholar]