Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Expert Review of Neurotherapeutics

ISSN: 1473-7175 (Print) 1744-8360 (Online) Journal homepage: https://www.tandfonline.com/loi/iern20

Antihypertensive agents in Alzheimer’s disease:


beyond vascular protection

Thibaud Lebouvier, Yaohua Chen, Patrick Duriez, Florence Pasquier & Régis
Bordet

To cite this article: Thibaud Lebouvier, Yaohua Chen, Patrick Duriez, Florence Pasquier & Régis
Bordet (2019): Antihypertensive agents in Alzheimer’s disease: beyond vascular protection, Expert
Review of Neurotherapeutics, DOI: 10.1080/14737175.2020.1708195

To link to this article: https://doi.org/10.1080/14737175.2020.1708195

Accepted author version posted online: 23


Dec 2019.
Published online: 27 Dec 2019.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iern20
EXPERT REVIEW OF NEUROTHERAPEUTICS
https://doi.org/10.1080/14737175.2020.1708195

REVIEW

Antihypertensive agents in Alzheimer’s disease: beyond vascular protection


Thibaud Lebouviera,b*, Yaohua Chen b,c
*, Patrick Duriezc, Florence Pasquierb,c and Régis Bordetc
a
Inserm URM_S1172, University of Lille, Lille, France; bDISTALZ, University of Lille, Lille, France; cInserm, CHU Lille, University of Lille, Lille, France

ABSTRACT ARTICLE HISTORY


Introduction: Midlife hypertension has been consistently linked with increased risk of cognitive decline Received 2 October 2019
and Alzheimer’s disease (AD). Observational studies and randomized trials show that the use of Accepted 19 December 2019
antihypertensive therapy is associated with a lesser incidence or prevalence of cognitive impairment KEYWORDS
and dementia. However, whether antihypertensive agents specifically target the pathological process Alzheimer’s disease;
of AD remains elusive. pathology; vascular risk
Areas covered: This review of literature provides an update on the clinical and preclinical arguments factors; hypertension;
supporting anti-AD properties of antihypertensive drugs. The authors focused on validated all classes of antihypertensive agents;
antihypertensive treatments such as angiotensin-converting enzyme inhibitors (ACEi), angiotensin prevention; neuroprotection;
receptor blockers (ARB), calcium channel blockers (CCB), β-blockers, diuretics, neprilysin inhibitors, treatment
and other agents. Three main mechanisms can be advocated: action on the concurrent vascular
pathology, action on the vascular component of Alzheimer’s pathophysiology, and action on nonvas-
cular targets.
Expert opinion: In 2019, while there is no doubt that hypertension should be treated in primary
prevention of vascular disease and in secondary prevention of stroke and mixed dementia, the place of
antihypertensive agents in the secondary prevention of ‘pure’ AD remains an outstanding question.

1. Background white matter lesion, and smaller brain volumes [9]. Many stu-
dies have tried to answer the question of whether antihyper-
The total number of people with dementia worldwide in 2018
tensive agents can prevent or slow down AD. There is a wealth
is estimated at 50 million and is projected to nearly triple,
of observational studies showing that the use of antihyperten-
reaching 150 million in 2050 [1]. Around 70% of these cases
sive therapy is associated with a lesser incidence or prevalence
are attributed to Alzheimer’s disease (AD), a devastating con-
of cognitive impairment and dementia [10–16]. With the excep-
dition leading to dependence to caregivers [2]. AD thus repre-
tion of the Rotterdam Study cohort, where the decrease of the
sents an enormous strain on the health-care system,
relative risk for AD was not significant [12], most studies yielded
underlining the need for medicines that would slow progres-
the same results when separating AD from all-type dementia
sion, halt or prevent AD.
[10,12–16]. Four pivotal prospective observational studies were
Findings from observational studies have linked several
performed in population-based cohorts [13,14,16,17].
vascular risk factors (VRFs) with increased risk of late-life cog-
Interestingly, the Honolulu-Asia Aging Study and the
nitive impairment and AD (reviewed in [3]). According to
Rotterdam Study showed a positive correlation between the
a recent meta-analysis, a third of AD cases worldwide could
reduction of AD risk and duration of exposure to antihyperten-
be attributed to seven modifiable factors (low education, mid-
sive drugs, supporting a mechanistic link between antihyper-
life hypertension, midlife obesity, diabetes, physical inactivity,
tensive drug use and AD prevention [14,16].
smoking, and depression) [4]. In a longitudinal 20 year-long
In an effort to circumvent the biases of observational stu-
study gathering 1,409 middle-aged individuals, future demen-
dies, at least three randomized trials were performed to exam-
tia (fulfilling AD criteria in 80% of cases) was significantly
ine whether antihypertensive treatment prevents dementia
predicted by high age, low education, hypertension, hyperch-
± AD occurrence [18–20]. Two large placebo-controlled trials
olesterolemia, and obesity [5]. In this context, the idea that
testing different antihypertensive regimens in elderly patients
VRF treatment may also interfere with AD pathological process
with hypertension had to be stopped because of interim
was put forth [6,7].
analysis showing a reduction in stroke or mortality [18,20].
Among VRFs, hypertension, diabetes, and dyslipidemia are
Despite this premature interruption, the SYST-EUR trial, includ-
readily modifiable with established therapeutic strategies [8].
ing 2418 patients ≥60 years [18], showed a marginally signifi-
But midlife hypertension is generally viewed as one of the main
cant decrease in the incidence of dementia after a median
evitable VRF for cognitive decline and AD [2,6,7,9,10]. Midlife
follow-up of 2 years (21 cases in the placebo group, 15 of
high blood pressure (BP) was also associated with an increase of

CONTACT Prof. Régis Bordet Regis.Bordet@univ-lille.fr Faculté de Médecine, pôle recherche Département de Pharmacologie Médicale 1, Université Lille 2,
place de Verdun, Lille 59045, France
*
These authors contributed equally to this work
© 2019 Informa UK Limited, trading as Taylor & Francis Group
2 T. LEBOUVIER ET AL.

models and postmortem studies largely documented


Article highlights a synergistic association between hypertension and the two
● Observational studies showed that the use of antihypertensive ther-
pathological hallmarks of AD, namely β-amyloid deposits and
apy is associated with a lesser incidence or prevalence of cognitive neurofibrillary tangles, suggesting that antihypertensive
impairment and dementia, whereas randomized trials and data on agents may also reverse AD pathophysiology [30–33].
established AD are issuable.
● Action on the concurrent vascular pathology is probably the main
This survey thus aims to provide an update on the clinical
cause of cognitive decline reduction by antihypertensive drugs. and preclinical arguments supporting anti-AD properties of
● Action on the vascular component of Alzheimer’s pathophysiology is antihypertensive drugs, and to review putative mechanisms
the most appealing of the possible modes of action of antihyperten-
sive drugs; several ARBs and CCBs demonstrated positive effects on
(Table 1). Our aims are to provide a comprehensive view of
vascular amyloid clearance and alterations of vascular function in AD a timely debate, and to inspire future therapeutic guidelines
models. and trials.
● Action on nonvascular targets could explain anti-AD properties of
selective antihypertensive drugs, such as CCBs with high brain pene-
tration or some ARBs with PPARγ agonist properties. Off-targets may
be detrimental as well, as was shown with ACE and neprilysin
2. Search strategy
inhibitors – even though their theoretical pro-amyloidogenic proper-
ties do not seem to be clinically meaningful.
We searched the PubMed database for relevant articles pub-
● From this review of literature, ARBs (telmisartan in particular) and lished between 1 January 2000 and 31 August 2019. Search
CCBs (nilvadipine in particular) may offer the most benefit by inter- terms were ‘Alzheimer’s Disease’ OR Alzheimer OR ‘Amyloid
fering in amyloid formation, breakdown and clearance, and also
improving endothelial function.
beta-Peptides’ AND ‘antihypertensive agents’ OR different
class of antihypertensive agents.
We searched the clinical studies and preclinical research
articles. For conciseness, we focused on validated treatments
only. The search was limited to articles in English and full
which were AD, 11 cases in the treatment group, 8 of which papers. Abstracts were reviewed and articles potentially meet-
were AD), a result that was confirmed by an extended open- ing inclusion criteria identified. Reference lists and reviews
label follow-up of 5 years [21]. The more recent HYVET trial were hand-searched.
included 3336 significantly older patients (≥80 years). After
follow-up of 2 years, there was no significant difference in
3. Angiotensin-converting enzyme inhibitors
the rate of incident dementia between treatment and placebo
groups despite a trend toward less incident AD (78 vs. 86 cases Angiotensin-Converting Enzyme (ACE) inhibitors are the oldest
in the placebo group). When these data were combined in class of drugs targeting the renin-aldosterone angiotensin
a meta-analysis with other placebo-controlled trials of antihy- system (RAAS), and one of the most prescribed antihyperten-
pertensive treatment, the combined risk ratio favored treat- sive agents. They act by binding to the active site of ACE and
ment [20]. Lately, the SPRINT-MIND trial showed that an interfering with the ability of the enzyme to bind to and
intensive BP control compared with standard practices did cleave its substrates. ACE cleaves the biologically inactive
not significantly reduce the incidence of probable dementia angiotensin I into the vasoconstrictive angiotensin II [34]. It
[22]. No attempt to classify dementia subtype was made. also transforms the vasodilator bradykinin into inactive
However, the authors showed a lower risk of incident mild peptides.
cognitive impairment in the intensive treatment arm (14.6 vs
18.3 cases per 1000 person-years). Moreover, an intensive BP
3.1. Preclinical studies
control was significantly associated with a smaller increase in
white matter lesion volume and a greater decrease in total The case for ACE inhibitors in AD became certainly weaker
brain volume [23]. Again, the trial was terminated earlier after a demonstration that ACE was an Aβ peptide-cleaving
because of a benefit on cardiovascular event. enzyme, which was first shown in cellular models [35] and
Data in established AD are scarcer. Some observational later demonstrated at a molecular level [36]. ACE converts
studies show a slower cognitive decline in patients treated Aβ43 and Aβ42 to the less aggregable Aβ41 and Aβ40 species,
with antihypertensive drugs [24,25], although it is not respectively, and its inhibition seems to enhance Aβ deposi-
a universal finding [26]. tion in the brain of amyloid-beta precursor (APP) transgenic
The putative protective effect of antihypertensive therapies mice [27,28]. Conversely, mice overexpressing ACE in mono-
against AD is thus a topic of increasing interest. However, cytes and macrophages display a reduction of Aβ deposits and
antihypertensive agents are the lead stroke-preventing treat- a retention of cognitive abilities [37]. This apparent protection
ment. They contribute to protect the cerebrovascular structure from AD pathology supports an important role for ACE in Aβ
from the macroscopic (e.g. white matter injury, hypoperfusion, clearance, at least in the context of a transgene overexpres-
microbleeds) and microscopic (e.g. blood–brain barrier disrup- sion, and claimed against the use of centrally active ACE
tion, endothelial dysfunction, microvessel rarefaction) deleter- inhibitors in AD.
ious effects induced by hypertension [27]. Although vascular However, the deleterious effects of ACE inhibitors were not
dysfunction is viewed as a major contributor to AD [28,29], it confirmed with two other APP transgenic lines [38].
proves extremely difficult to distinguish a possible effect Normalization of the excessive hippocampal ACE activity was
on AD pathophysiology from the known effect of antihyper- even beneficial to Tg2576 APP mice in one study, by decreas-
tensive agents on the vascular burden in the brain. Yet animal ing amyloidogenic processing of APP and formation of
EXPERT REVIEW OF NEUROTHERAPEUTICS 3

Table 1. Effect of antihypertensive drugs on Alzheimer’s pathophysiology.


Antihypertensive drugs Preclinical studies Clinical trials Global outcome
Angiotensine-converting Absence of noxiousness
enzyme inhibitors Not sufficient data to support
a neuroprotective effect
Captopril ↓ Amyloidogenic processing ([39,41])
↓ ROS [39]
↓ Inflammation [41]
Perindopril ↓ ACE activity [40] No effect [19,43]
Angiotensin receptor Encouraging positive preclinical data
blockers Potential candidate for future trial, especially
telmisartan
Valsartan ↓ Aβ deposit [51,52]
↓ Aβ oligomerization [51,52]
Losartan ↓ Aβ deposit [51,52]
↓ Aβ oligomerization [51,52]
↓ Tau phosphorylation [59]
↓ Oxidative stress in microvessels [61]
Candesartan ↓ Aβ oligomerization [51,52] Slight effect in some cognitive domains
[66]
Telmisartan ↓ LDL-R and ApoE [55]
↑ Cerebral blood flow [53]
↓ Expression of cytokines [54,57]
↓ Glial activation [58]
Calcium channel blockers Encouraging data for a future trial, with
more selective criteria of patients
Nimodipine ↓ Calcium release [79–81] Slight effect on cognitive function [91]
↓ Amyloidogenesis [82]
↓ Expression of cytokines [88]
↓ Iron entry and toxicity [89]
Nilvadipine ↓ Amyloidogenesis [78] No effect on cognitive outcome, except for
↑ Aβ42 clearance [78,85] male APOE4 [97]
Inhibition of vasoactivity and ↑ Regional cerebral blood flow [96]
restoration of cortical perfusion [86]
Nitrendipine ↓ Amyloidogenesis [84] ↓ Incidence of dementia by 50%, especially
↓ Tau phosphorylation [84] for AD (8 vs. 15 cases) [21]
Nicardipine ↓ Iron entry and toxicity [90]
β-Blockers ↓ Hippocampal Aβ42 and [99–102] Contradictory results in cohorts, no No sufficient data to support
↓ Hyperphosphorylated tau [99–101] available clinical trial [11,105] a neuroprotective effect
Diuretics Not sufficient preclinical data, mostly results
from cohorts
Thiazide diuretics ↓ Aβ oligomerization [51] None
Potassium-sparing diuretics Raise and maintain potassium level Trend to lesser incidence [20]
In cohorts, reduction of risk to develop AD
[17,106]
Neprilysin inhibitors ↑ Aβ levels [110,111] Low brain penetration [114] Not clarified for the absence of noxiousness
α-1-Adrenoceptor Release of APOE and cytokines [115] Improve behavioral symptoms [116] Not clarified for the neuroprotective effect
antagonist (prazosin)

reactive oxygen species [39]. In a rat model of intra- [42], a finding that was not replicated in other cohorts.
hippocampal injections of Aβ, perindopril improved learning Perindopril was not superior nor inferior to placebo to prevent
and memory, which correlated with decreased ACE activity incidence of non-stroke-related dementia [19] or continued
and delayed AD pathology [40]. Furthermore, ACE inhibitors cognitive decline [43] in the Perindopril Protection Against
might be beneficial by reducing glial-induced inflammation, Recurrent Stroke Study (PROGRESS). In two prospective obser-
since angiotensin II leads to unregulated inflammation by glial vational studies, centrally active ACE-inhibitors like perindopril
activation and to inhibition of acetylcholine release. A recent or captopril seemed to prevent cognitive decline in both
study demonstrated in vitro anti-inflammatory properties of cognitively normal [44] and demented patients [45], as com-
captopril, and reduced amyloid burden, macrophage/micro- pared with non-centrally active ACE inhibitors that do not
glia accumulation by an intranasal captopril treatment as cross the blood–brain barrier. To further address putative pro-
well [41]. amyloidogenic effects of ACE inhibitors, a cerebrospinal fluid
(CSF) study showed that ramipril inhibited ACE activity in the
CSF and improved blood pressure, but did not influence CSF
3.2. Clinical trials
Aβ1-42 [46]. One interesting pharmacogenetics study showed
Human studies testing ACE inhibitors support the absence of that the use of ACE-inhibitors during 1 year might slow the
noxiousness of ACE inhibitors. Only one observational study in cognitive decline in patients with AD, but only for APOE4
a cohort of >6,000 demented patients in UK showed an asso- carriers of specific ACE genotypes [47].
ciation between ACE-inhibitors use and a slight increase risk of Overall, if some caution was justified regarding the use of
mortality in patients with AD (adjusted HR 1.19 [95% CI ACE inhibitors in AD, available preclinical data are inconclusive
1.07;1.33] but without evidence of increased hospitalization and clinical data cleared most concerns regarding their
4 T. LEBOUVIER ET AL.

experimental pro-amyloidogenic properties. This is probably ischemia [57]. More recently, telmisartan was shown to reduce
consistent with the much lower efficiency of hydrolysis of Aβ glial activation [58] in the 5XFAD transgenic mouse model.
by ACE compared to other Aβ degrading enzymes, such as ARBs could also interfere with the AD process through tau-
neprilysin, endothelin-converting enzyme, and insulin- mediated effects: in the rat, intracerebroventricular injections
degrading enzyme (reviewed in [48]). ACE inhibitors might of angiotensin II significantly increased tau phosphorylation,
also act by other pathways which are beneficial for cognition, an effect that was attenuated by losartan [59]. The relevance
as shown in some preclinical studies above. As we will later of those findings for AD however remains doubtful, since tau
discuss, a similar debate was recently stirred up by the new phosphorylation is a nonspecific answer to any kind of neuro-
class of neprilysin inhibitors. nal insult. In line with this, losartan-induced prolonged hypo-
tension was also shown to lead to tau hyperphosphorylation
through oxidative stress [50].
4. Angiotensin receptor blockers Beyond the effects on AD pathology, two remarkable
studies yielded the same interesting results using different
Angiotensin receptor blockers (ARBs) act downstream of ACE
ARBs (olmesartan and losartan) and different treatment
inhibitors in the RAAS. The ARBs are non-peptide compounds
schemes on different APP transgenic mouse lines. Both
that specifically block the binding of angiotensin II to the
demonstrated that ARBs protected cognitive function and
angiotensin I receptor [34].
cerebrovascular activity through decreased oxidative stress
in brain microvessels. Interestingly, those benefits occurred
without any decrease in Aβ levels and were not related to
4.1. Preclinical studies
changes in blood pressure [60,61].
Most preclinical studies generally support the use of ARBs
in AD, with only rare exceptions [49,50]. The work of The
4.2. Clinical trials
Mount Sinaï group in New York stands among the interesting
data published on ARBs in preclinical AD models [51]. By Observational studies on ARBs are conflicting. While
screening 55 commonly prescribed antihypertensive drugs a population-based cohort study showed that ARB treatment
looking for anti-Aβ properties in vitro, they determined that was associated with a reduced risk of dementia, including AD
only valsartan and losartan were capable of both lowering Aβ [62], those results were not confirmed in a cohort of hyper-
and decreasing oligomerization of Aβ peptides in primary tensive patients [63], raising the possibility that hypertension
neuronal cultures, while candesartan acted on oligomerization was a confounding factor in the first study.
only [51,52]. Going to animal models, they showed that pre- More interesting is the SCOPE trial, which included 4937
ventive treatment of Tg2576 mice with valsartan significantly patients aged 70 to 89 years with mild-to-moderate hyperten-
reduced brain Aβ deposits and Aβ-mediated cognitive dete- sion and Mini-Mental State Examination (MMSE) score ≥24.
rioration, independently of blood-pressure lowering [52]. Double-blind treatment was initiated with candesartan or pla-
Unfortunately, those anti-amyloidogenic properties did not cebo, and an open-label therapy was added as needed to
appear to be specific to ARBs as a class, since other ARBs, like control BP. While there was no difference in the whole popu-
telmisartan, did not have any in vitro effect on Aβ, and there are lation of the study, the MMSE score declined less in the low
no available data in vivo. In an acute mouse model of intracer- cognitive function group (MMSE 24–28) receiving candesartan
ebroventricular injection of Aβ, telmisartan was able to reduce [64] (reviewed in [65]). This analysis was not pre-specified but
amyloid deposition [53] and improve cognition [53,54], thereby suggested that candesartan was able to hamper an initiated
suggesting that mechanisms other than action on Aβ produc- cognitive decline, regardless of its degenerative or vascular
tion or aggregation may decrease the amyloid load. Actually, nature. In a cognitive sub-study performed in 257 subjects
telmisartan draws an increasing interest because of a putative enrolled in SCOPE, the candesartan group showed less decline
pleiotropic vascular protection. Telmisartan is indeed a partial on attention and episodic memory but no difference in execu-
agonist of peroxisome proliferator-activated receptor-γ (PPARγ), tive function and working memory [66]. Although caution
a central regulator in insulin and glucose metabolism. should be exerted, the cognitive domains on which candesar-
Interestingly, in spontaneously hypertensive stroke resistant tan had a protective effect do not discard an effect on an AD
rats, chronically administered telmisartan strongly reduced low- (rather than vascular) component.
density lipoprotein receptor (LDL-R) and ApoE expression in We did not find published clinical studies on ARBs in AD,
neurons [55]. Since ApoE and LDL-R are key elements in the underlining the gap between a wealth of encouraging precli-
amyloidogenic processing of APP [56], these data provide nical data and the paucity of clinical trials in AD. Few clinical
a potential mechanism for the possible anti-amyloidogenic trials are ongoing [67,68], using mostly telmisartan and losar-
properties of telmisartan. In a simpler model of intracerebral tan, with an estimated study completion in 2021.
injection of Aβ, telmisartan also enhanced cerebral blood flow Overall, while telmisartan has certainly an interesting pro-
and attenuated the Aβ-induced increase in expression of cyto- file, and PPARγ is a longstanding target in AD [69], its super-
kines [53,54]. The smaller efficacy of losartan and the partial iority to other ARBs remains questionable. Indeed, many ARBs
reversion of the effects by a PPARγ antagonist supported an share to some extent some PPARγ agonist properties [70] and
added value for telmisartan [53,54]. Those beneficial effects on the studies performed with valsartan, losartan, and olmisartan
cognition and cytokine release were confirmed in a rat model on AD transgenic mice provide more compelling evidence
combining intracerebroventricular injection of Aβ and cerebral than the ones performed with telmisartan on short-term Aβ
EXPERT REVIEW OF NEUROTHERAPEUTICS 5

injection models. Beyond the effect on Aβ pathology, which that nilvadipine treatment increased plasma Aβ level in trans-
does not seem to be shared by all ARBs, the most interesting genic animals, Paris et al. showed that some CCBs, among
and consistent data are the ones addressing their positive which nilvadipine, were able to increase Aβ42 transcytosis
effects on vasculature and cognition. Interestingly in that across an in vitro blood–brain barrier model, as well as to
regard, a confidential positron electron tomography study increase brain clearance of intracranially administered human
showed that telmisartan as compared to nimodipine increased Aβ42 [78,85]. Aβ clearance properties were independent of
rCBF in 10 AD patients after 6 month-treatment [71]. blood-pressure lowering, brain penetration of the drugs and
L-type CCB inhibition, suggesting that nilvadipine acted
directly on brain endothelial and/or mural cells [84]. Beyond
5. Calcium channel blockers
its effect on Aβ transcytosis, nilvadipine was shown to inhibit
Calcium ions are critical mediators of cell signaling, and cal- the vasoactivity elicited by Aβ in isolated human arteries and
cium channels are ubiquitously expressed. Far from being to restore cortical perfusion levels in transgenic mice [86].
specific to vascular smooth muscle cells, the L-type channels, Among the additional possible mechanisms of neuropro-
the primary target of calcium channel blockers (CCBs), are tection by CCBs are inhibition of Tau hyperphosphorylation
heavily expressed by neurons [72,73] and may be reached by [84,87], inhibition of Aβ-induced release of proinflammatory
CCBs depending on brain penetration. Hence, CCBs are good cytokines by microglia [88], and prevention of iron entry
candidates as pleiotropic drugs that could influence both and toxicity [89,90].
neural conduction and cerebral blood flow to circumvent AD.
The effect of CCBs in AD models has been studied since the
5.2. Clinical studies
1990s [74]. Although a few studies have focused on verapamil
and diltiazem [75], most have been performed with CCBs of The dementia prevention trials performed with CCBs were
the dihydropyridine family. Nimodipine is the most studied of pioneered by the SYST-EUR vascular dementia sub-study,
this class, because its lipophilic properties favor brain penetra- which investigated whether a long-term treatment with
tion. Recently, however, interesting data have emerged on nitrendipine could reduce the incidence of dementia [18]. As
nilvadipine. Both drugs block L-type calcium channels, but mentioned earlier, the SYST-EUR study was prematurely inter-
nimodipine also acts on T-type channels [72,73]. rupted after an interim analysis showing a 42% reduction in
the incidence of stroke in the treated group. However, 2,418
patients with a median follow-up of 2 years participated to the
5.1. Preclinical studies
sub-study. Active treatment reduced the incidence of demen-
Most published work supports an action of CCBs on amyloid. tia by 50% (11 vs. 21 cases), and those results were confirmed
In rats, impairment of spatial memory following intracerebro- in the open-label extension study [21]. Unexpectedly, nitren-
ventricular injection of Aβ was prevented both by nilvadipine dipine prevented more Alzheimer’s dementia (8 vs. 15 cases)
and nimodipine treatments [76,77]. In a thorough study, Paris than vascular dementia (3 vs. 6 cases). The criteria used to
et al. showed that chronic oral treatment with nilvadipine define vascular and Alzheimer dementias were however ques-
decreased Aβ burden in the brains of transgenic PS1/APPsw tionable, even at that time, and one can wonder whether AD
mice, and improved learning abilities and spatial memory [78]. diagnosis would have withstood the use of brain MRI and
The putative mechanisms of action of CCBs on amyloid are biomarkers.
threefold. The first hypothesis is that CCBs suppress Aβ- Nimodipine has been extensively tested in dementia
induced calcium release. Indeed, in vitro studies suggest that patients in the 1990s. The drug has even had the privilege of
the neurotoxicity of Aβ species is mediated by an increase in a 2005 Cochrane Review to evaluate its effects on dementia.
cytosolic calcium, resulting from direct or indirect stimulation Of the 14 trials included in the meta-analysis, 2 trials included
of L-type calcium channels [79]. Nimodipine was repeatedly only patients with AD, 9 trials included only patients with
shown to attenuate Aβ-induced neurotoxicity [80,81]. cerebrovascular dementia (CVD), and three trials included
Inhibition of amyloidogenesis is the second putative effect patients with AD, CVD, and mixed disease. In hindsight, inclu-
of CCBs. In primary cultures expressing human APP, sion criteria are now obsolete, judgment criteria are only
a sustained high concentration of cytosolic calcium inhibited clinical and study durations are too short. However,
the alpha-secretase cleavage of APP (non-amyloidogenic path- a beneficial effect was seen on cognitive function and on
way) and induced the production of intraneuronal Aβ42 (amy- overall clinical improvement at 12 weeks with a dose of
loidogenic pathway), a process that was prevented by 90 mg/day. There was no effect on functional scales [91].
nimodipine [82]. The latter is controversial since nimodipine With the emergence of cholinesterase inhibitors, clinical
was shown to increase Aβ42 production and secretion in research on nimodipine was dropped in the 2000s despite
a different culture setting [78,83]. Paris et al. showed that these encouraging results in short-term clinical trials. Only
among several L-type CCBs, only nilvadipine and to a lesser one small 24-week trial showed no advantage of the combina-
extent nitrendipine were able to lower Aβ production, inde- tion of galantamine/nimodipine over galantamine alone [92].
pendently of their action on L-type calcium channels. This Following exciting preclinical results, the rather old drug
effect might be due to inhibition of the spleen tyrosine kinase nilvadipine has recently gained its way to clinical trials. In
Syk, which indirectly inhibits the β-cleavage of APP [78,84]. a proof-of-concept case report of a mild AD patient, nilvadi-
The third and perhaps most interesting mechanism of pine elevated both the regional cerebral blood flow and the
action of CCBs is on brain vasculature. From the observation Mini-Mental State Examination (MMSE) score [93].
6 T. LEBOUVIER ET AL.

Recapitulating the beneficial short-term effects observed 6.2. Diuretics


with nimodipine, a six-week, open-label study gathering 55
There are few preclinical reports on diuretics in AD models.
patients with mild to moderate AD showed that nivaldipine
The aforementioned high throughput screening of drugs able
was well tolerated and there was a treatment benefit on the
to inhibit oligomerization Aβ42 showed that furosemide did
MMSE and executive functions [94,95]. The recent 18-month
not only prevent oligomerisation in vitro but also dissociated
European multicentre NILVAD trial was designed to demon-
oligomers in the brain of Tg2576 mice [51]. Research on
strate a positive effect of nilvadipine, based on this encoura-
furosemide did not go further to date.
ging data. The study enrolled more than 500 people with
Clinical trials on diuretics are mostly represented by preven-
mild to moderate AD according to the 1984 NINCDS-ADRDA
tion trials. The aforementioned negative HYVET trial used the
criteria, and compared a once-daily dose of 8 mg nilvadipine
potassium-sparing diuretic indapamide as the first-line treat-
to placebo during 78 weeks [96]. The study failed to demon-
ment, combined with 2 to 4 mg perindopril if needed [20].
strate any effect of nilvadipine on the cognitive outcome.
There was however a trend toward a lesser incidence of AD in
However, in predefined subgroup analyses, there was less
the treated group. Likewise, in the Ginkgo Evaluation in
cognitive decline in patients with MMSE >20, in males and in
Memory Study (GEMS), cognitively normal persons on diuretics,
APOE ε4 carriers [97]. As found in the aforementioned case-
ACE inhibitors or ARBs and mild cognitive impairment patients
report, a subgroup of patients in the NILVAD trial showed an
on diuretics had a 50% reduction in the risk of developing
increase of cerebral blood flow in hippocampal regions [98].
dementia [106]. Last, the 3-year reduction in AD risk observed
Overall, CCBs show preclinical arguments favoring
with the antihypertensive treatment in the Cache County study
a protective role against AD. This putative role does not appear
was greatest with the diuretics, and specifically potassium-
to be a drug class-effect. Only selected molecules within this
sparing diuretics [17].
class yielded interesting results – perhaps depending on brain
penetration. The clinical outcome of CCBs are more mitigated,
and possibly specific subgroup of patients should be targeted. 6.3. Neprilysin inhibitors
Neprilysin is a ubiquitous neutral endopeptidase. Peripherally,
6. Other antihypertensive agents its inhibition increases the bioavailability of natriuretic peptides,
The other classes of antihypertensive agents have brought bradykinin, and substance P, resulting in a strong antihyperten-
comparatively less attention. This relative paucity of data is sive effect through increased natriuresis and vasodilation. The
due to deceptive preliminary data, absence of brain penetra- first-in-class neprilysin inhibitor sacubitril was successfully tested
tion or (in the case of neprilysin inhibitors) novelty of the in hypertension [107] and heart failure [108] in combination with
drug. the ARB enalapril. To date, enalapril/sacubitril combination has
only been approved for heart-failure patients.
This success-story was tarnished by a warning issued in
6.1. β-Blockers 2015 [109]. Neprilysin is indeed an Aβ-degrading enzyme.
The nonselective β-blocker propanolol has been the subject of While it is not clear to what extent neprilysin contributes to
a few encouraging reports from a single group, showing Aβ clearance in humans, preclinical data suggest that neprilysin
a reduction of cognitive impairment and hippocampal Aβ42 deficiency increases brain Aβ levels [110] and worsens cognitive
and hyperphosphorylated tau levels in propranolol-treated deficits and Aβ pathology in the 5XFAD transgenic mouse
Tg2576 mice [99]. Interestingly the results were recapitulated model [111]. Hence, the same questions that surrounded ACE
in senescence-prone mice spontaneously [100] or chronic cor- inhibitors years ago are currently stirring up a controversy on
ticosterone administration [101]. Comparatively, the selective neprilysin inhibitors [109,112]. Reassuring preliminary results
β1 adrenergic receptor antagonist nebivolol significantly came recently. In a thorough study performed on cynomolgus
reduced brain amyloid content but failed to improve cognitive monkeys, a well-known AD model that spontaneously develops
function [102]. However, the use of centrally acting β-blockers brain amyloidosis, there was a significant short-term increase in
is hindered by the possible involvement of beta-adrenergic Aβ species half-life and cerebrospinal fluid levels, but levels
systems in learning and memory [103,104]. normalized after 2 weeks [113]. A 39-week treatment did not
Clinical studies on β-blockers are confusing. In 2,212 affect amyloid pathology [113]. Both studies were sponsored by
African-American adults aged 65 years and older, centrally the pharmaceutical company that develops sacubitril. In
acting sympatholytic agents, as opposed to other antihyper- humans, brain penetration of the active metabolites of sacubi-
tensive agents, were associated with an increased risk of tril was low and the drug did not change cerebrospinal fluid
diagnosis of cognitive impairment [11]. In sharp contrast levels of Aβ40 and Aβ42 [114].
are the results from the Honolulu-Asia Aging Study, which
prospectively followed 2,197 elderly Japanese American men
6.4. α-1-Adrenoceptor antagonist and other alkaloid
during more than 15 years. In this cohort, β-blocker use as
drugs
the sole antihypertensive drug at baseline was associated
with a lower risk of cognitive impairment as compared with With the rationale that enhanced responsiveness to norepi-
men not taking any antihypertensive medications. This asso- nephrine at the α-1-adrenoceptor may contribute to the
ciation was not found with other antihypertensive pathophysiology of dementia, the centrally-acting α-1-adreno-
agents [105]. ceptor antagonist prazosin was tested in vitro and in vivo.
EXPERT REVIEW OF NEUROTHERAPEUTICS 7

Prazosin was able to reduce the generation of amyloid β in and cerebrovascular disease is something more than the mere
neuroblastoma cells but did not influence amyloid plaque addition of two highly prevalent conditions. Concurrent cerebro-
load in transgenic APP23 mice. However, prazosin seemed to vascular disease is more common in AD than in any other
prevent memory deficits over time, possibly through neurodegenerative disease [122], occurring in at least one-third
the release of apolipoprotein E and anti-inflammatory cyto- of all cases [123]. Vascular and AD pathologies may act synergis-
kines [115]. Clinically, prazosin was tested in a small double- tically to increase the likelihood of neuronal death – and hence
blind, placebo-controlled, parallel group study. The drug was symptoms – in dementia of mixed vascular and neurodegenera-
well tolerated and improved behavioral symptoms in patients tive etiology [122]. According to the neurovascular hypothesis
with agitation/aggression in AD [116]. Neuroprotective effects of AD, brain vascular insults might even be causative in most
were not tested. late-onset sporadic forms of AD (reviewed in [124]). Incident
Two small reports additionally showed that Reserpine, vascular pathology is indeed associated with a faster decline
a pleiotropic indole alkaloid drug that has been used both as in AD [125,126].
an antipsychotic and antihypertensive agent, prevents Aβ toxi- Logically, preventing overt strokes or silent microvascular
city and increases lifespan in a C. elegans AD model [117,118]. Its lesions will have a positive effect on the cognitive outcome. In
numerous side-effects probably dissuade further research in AD. that regard, antihypertensive drugs participate to reduce the
vascular burden in AD. The PROGRESS study clearly shows that
an antihypertensive agent like perindopril protects against
7. Discussion
post-recurrent stroke dementia, and not against dementia
Several mechanisms can be advocated when trying to under- without recurrent stroke [19]. Once a putative neurodegenera-
stand how antihypertensive agents could slow down AD pro- tive process has engaged, perindopril seems to be less effec-
gression (Figure 1). Putative mechanisms are threefold: (1) tive [43], and any remaining activity of perindopril could be
action on the concurrent vascular pathology; (2) action on due to the prevention of silent strokes [120].
the vascular component of Alzheimer’s pathophysiology; (3) On the other hand, the action of antihypertensive agents
action on a nonvascular component. might go well beyond the prevention of vascular lesions. In
the Honolulu-Asia Aging Study, antihypertensive agents abol-
ish the positive association between hypertension and hippo-
7.1. Action on the concurrent vascular pathology
campal atrophy, which is one of the radiological hallmarks
Hypertension is one of the long-known risk factors for stroke of AD [127]. Furthermore, in a postmortem study of 291 brains
[8,119] or silent cerebrovascular disease [120,121]. While the link with normal brain aging or AD pathology, but no significant
between VRFs and AD only begins to be ascertained, the relation cerebrovascular disease, there was substantially less AD neu-
between AD and cerebrovascular disease has been extensively ropathology in the medicated hypertension group than in the
studied in recent years [28]. AD is generally not seen as a primary non-medicated hypertension and non-hypertension
vascular disease. However, although mechanistic links remain groups [128]. Thus, there are hints, but no hard evidence
elusive, it is becoming obvious that the concomitance of AD from clinical studies supporting a protective effect of

Figure 1. Putative mechanisms of action of antihypertensive drugs on Alzheimer’s disease.


8 T. LEBOUVIER ET AL.

antihypertensive drugs toward AD beyond the prevention of Alternatively, drugs may act on one or several secondary
cerebrovascular pathology. targets unrelated with the one responsible for its antihyperten-
sive effect. The PPARγ agonist properties of several ARBs [70]
fall in that category. Likewise, the possible superiority of nilva-
7.2. Action on the vascular component of Alzheimer’s dipine to other CCBs in preclinical studies might be due to
pathophysiology targets other than L-type calcium channels, such as the tyrosine
kinase Syk [84].
Preventing cerebrovascular events may have indirect conse-
Interestingly, unexpected effects may as well disfavor the
quences on the AD process, since there are putative pathophy-
drug, as showed the controversies surrounding ACE and nepri-
siological links between ischemic injury, aberrant
lysin inhibitors, which were both suspected of promoting AD
beta-amyloid processing and neuronal dysfunction [124].
pathology by inhibiting Aβ-degrading enzymes.
Above all, the treatment of VRFs could act upstream of
a shared pathway leading to vascular pathology on the one
hand and to AD on the other [124]. Alteration of the neurovas- 8. Expert opinion
cular unit and the blood–brain barrier are thought to play
Whether and how the treatment of hypertension (and VRFs as
a central role in AD pathophysiology and symptoms [124]. In
a whole) hampers Alzheimer’s pathological process is a timely
most AD cases, β-amyloid deposits also occur around capillaries
issue. While many observational and interventional studies
and in arterial walls (a condition termed cerebral amyloid angio-
suggest that improved control of hypertension decreases
pathy) [129,130]. By causing collagenous thickening and reduced
dementia incidence, it remains doubtful whether antihyper-
pulsatility of arteries and arterioles, hypertension could in turn
tensive drugs prevent AD specifically. Definitive proof will be
impair perivascular drainage of β-amyloid and prompt cerebral
hard (even impossible?) to obtain since dementia prevention
amyloid angiopathy [131]. Vascular pathology also causes
trials in hypertensive individuals are now unethical, consider-
a blood–brain barrier breakdown which may also accelerate
ing that hypertension decreases the risk of cardiovascular and
amyloidogenesis, and decrease brain perfusion [132]. Be they
cerebrovascular events – even in the oldest old [20]. The place
the cause or consequence of AD, preserving blood–brain barrier
of antihypertensive agents in the secondary prevention of AD
integrity and neurovascular unit function might both favor the
is a simple question – yet remains an area of uncertainty.
vascular clearance of Aβ and preserve neurovascular coupling,
Research is desperately needed to tackle the outstanding
whose demise is one of the earliest observable changes in AD.
question of the role of antihypertensive drugs in the primary
Antihypertensive drugs, and particularly the ARBs and the
prevention of AD. The relative paucity of preclinical research
CCBs are interesting in that regard, because preliminary data
studies and the lack of state-of-the-art clinical trials testing
show positive effects on vascular amyloid clearance [78,85]
a vascular intervention are striking. Until now, research fund-
and alterations of vascular function [51,52,58,59,84,96] in AD
ing was probably the main limiting factor, because most anti-
models. While those properties seem to be shared by several
hypertensive agents are in the public domain, and studies on
ARBs and may represent a class-effect, only individual drugs
VRFs depend mostly on academic funding. On the preclinical
in the CCB class yielded positive results, probably depending
side, the swiftness of the publication and the state-of-the-art
on brain penetration. Interestingly, beyond AD models, ARBs
techniques used in the sacubitril study recently demonstrated
attenuate vascular oxidative stress in chronic hypoperfu-
the quality shift of industry-funded work [113], that could
sion [133], and restore blood–brain barrier integrity in vitro
serve as a reference to reassess the effect of the main anti-
[134] and in vivo [135,136], in hypertensive encephalopathy
hypertensive drugs on amyloid processing. On the clinical
and diabetes models. Those protective effects result in
side, the use of CSF biomarkers and neuroimaging (3T-MRI
improved cognition independently of blood-pressure lower-
and tau/amyloid PET) is eagerly awaited to assess the
ing in an hypertensive rat model [135].
effect of selected long-term antihypertensive treatment on
Alzheimer pathology and vascular burdens.
For future trials, new diagnostic criteria [141,142] and neu-
7.3. Action on a nonvascular component of Alzheimer’s
roimaging markers [143,144] should be systematically used to
pathophysiology
better differentiate their respective action on the vascular load
Third, the possibility that drugs approved for hypertension may and on AD pathophysiology. Above all, future trials should
interact with AD-related pathophysiological pathways through answer the one question with real therapeutic implications:
mechanisms unrelated to its original therapeutic intention must should patients with AD but without major hypertension nor
not be discarded [137,138]. The concept of drug repositioning cerebrovascular disease be treated with the same stringent
in AD, beyond antihypertensive agents, has been discussed in objectives as in secondary prevention of stroke, despite poten-
recent reviews [139,140]. Antihypertensive agents may have tial side effects? A drop of BP is commonly observed several
unforeseen effects on Alzheimer’s pathological process because years before the onset of symptoms in late-onset dementia
of an extravascular, neuroglial expression of the primary target. [145], and BP continues to fall after the diagnosis [146]. The
The CCBs are probably the best example of such effects, since effect of antihypertensive drugs should thus be carefully mon-
calcium channels are also expressed by neurons. Calcium fluxes itored in dementia, and their overall use can be questioned,
are potentially involved in amyloidogenesis and neurodegen- especially in frail patients. Yet evidence of an increased risk of
eration, providing a strong rationale to their putative protective orthostatic hypotension in demented patients treated with
properties against AD. antihypertensive drugs is lacking. In the NILVAD study, there
EXPERT REVIEW OF NEUROTHERAPEUTICS 9

was no evidence of an increased risk of orthostatic hypoten- 2. Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer disease.
sion, fractures, falls, syncope, or dizziness in patients with Nat Rev Neurol. 2011;7:137–152.
3. O’Brien JT, Markus HS. Vascular risk factors and Alzheimer’s disease.
mild-to-moderate AD, with or without hypertension, treated BMC Med. 2014;12:218.
by 8 mg of nilvadipine during 78-weeks [147]. • A comprehensive review on relationship between vascular risk
Studies should include individuals with preclinical or early factors and Alzheimer’s disease.
prodromal AD and plan a long follow-up period. As the associa- 4. Norton S, Matthews FE, Barnes DE, et al. Potential for primary
tion between antihypertensive drugs and AD may depend on prevention of Alzheimer’s disease: an analysis of population-
based data. Lancet Neurol. 2014;13:788–794.
sex, ethnicity and genetic background (APOE status), future 5. Kivipelto M, Ngandu T, Laatikainen T, et al. Risk score for the prediction
therapeutic strategies will probably be personalized [148]. of dementia risk in 20 years among middle aged people: a longitudinal,
Recent evidence of the efficacy of polypills associating two population-based study. Lancet Neurol. 2006;5:735–741.
antihypertensive drugs (diuretic and ACE inhibitor), statin, and 6. Valenzuela M, Esler M, Ritchie K, et al. Antihypertensives for com-
aspirin came from a pragmatic trial in Iran, showing a reduction bating dementia? A perspective on candidate molecular mechan-
isms and population-based prevention. Transl Psychiatry. 2012;2:
of the risk of major cardiovascular events. These findings are e107.
particularly important for low-income and middle-income coun- 7. Safouris A, Psaltopoulou T, Sergentanis TN, et al. Vascular risk
tries and should be taken into account by the scientific commu- factors and Alzheimer’s disease pathogenesis: are conventional
nity in the conception of future trials. A combination of different pharmacological approaches protective for cognitive decline pro-
classes of antihypertensive treatment might be of interest both gression? CNS Neurol Disord Drug Targets. 2015;14:257–269.
8. Meschia JF, Bushnell C, Boden-Albala B, et al. Guidelines for the
at scientific and pragmatic levels, favoring an ‘efficient access’ to primary prevention of stroke: a statement for healthcare profes-
treatment [149]. Last, in the big data era, large-scale observa- sionals from the American Heart Association/American Stroke
tional data may further support a long-term effect of antihyper- Association. Stroke. 2014;45:3754–3832.
tensive drugs on cognition and AD. 9. Lane CA, Barnes J, Nicholas JM, et al. Associations between blood
For our current practice, although the target blood-pressure pressure across adulthood and late-life brain structure and pathol-
ogy in the neuroscience substudy of the 1946 British birth cohort
level reduction remains to be defined, we, as others [3,150], (Insight 46): an epidemiological study. Lancet Neurol.
deem reasonable to initiate an antihypertensive treatment in 2019;18:942–952.
hypertensive AD patients and in AD patients with evidence of 10. Tzourio C, Dufouil C, Ducimetière P, et al. Cognitive decline in
concomitant cerebrovascular disease. From this review of litera- individuals with high blood pressure: a longitudinal study in the
ture, ARBs (telmisartan in particular) and CCBs (nilvadipine in elderly. EVA study group. Epidemiology of vascular aging.
Neurology. 1999;53:1948–1952.
particular) may offer the most benefit by interfering in amyloid 11. Richards SS, Emsley CL, Roberts J, et al. The association between
formation, breakdown and clearance, and also improving vascular risk factor-mediating medications and cognition and
endothelial function. dementia diagnosis in a community-based sample of
African-Americans. J Am Geriatr Soc. 2000;48:1035–1041.
12. Ruitenberg A, Hofman A, Stricker BH, et al.; in’t Veld BA.
Funding Antihypertensive drugs and incidence of dementia: the
Rotterdam Study. NBA. 2001;22:407–412.
This paper was not funded. 13. Qiu C, Winblad B, Fastbom J, et al. Combined effects of APOE
genotype, blood pressure, and antihypertensive drug use on inci-
dent AD. Neurology. 2003;61:655–660.
Declaration of interest 14. Peila R, White LR, Masaki K, et al. Reducing the risk of dementia:
efficacy of long-term treatment of hypertension. Stroke.
The authors have no relevant affiliations or financial involvement with any 2006;37:1165–1170.
organization or entity with a financial interest in or financial conflict with 15. Hanon O, Pequignot R, Seux M-L, et al. Relationship between
the subject matter or materials discussed in the manuscript. This includes antihypertensive drug therapy and cognitive function in elderly
employment, consultancies, honoraria, stock ownership or options, expert hypertensive patients with memory complaints. J Hypertens.
testimony, grants or patents received or pending, or royalties. 2006;24:2101.
16. Haag MDM, Hofman A, Koudstaal PJ, et al. Duration of antihyper-
tensive drug use and risk of dementia: A prospective cohort study.
Reviewer Disclosures Neurology. 2009;72:1727–1734.
17. Khachaturian AS, Zandi PP, Lyketsos CG, et al. Antihypertensive
Peer reviewers on this manuscript have no relevant financial relationships medication use and incident Alzheimer disease: the Cache
or otherwise to disclose. County Study. Arch Neurol. 2006;63:686–692.
18. Forette F, Seux ML, Staessen JA, et al. Prevention of dementia in
randomised double-blind placebo-controlled Systolic Hypertension
ORCID in Europe (Syst-Eur) trial. Lancet. 1998;352:1347–1351.
19. Tzourio C, Anderson C, Chapman N, et al. Effects of blood pressure
Yaohua Chen http://orcid.org/0000-0003-1778-1790 lowering with perindopril and indapamide therapy on dementia
and cognitive decline in patients with cerebrovascular disease.
Arch Intern Med. 2003;163:1069–1075.
References 20. Peters R, Beckett N, Forette F, et al. Incident dementia and blood
pressure lowering in the Hypertension in the Very Elderly Trial
Papers of special note have been highlighted as either of interest (•) or of cognitive function assessment (HYVET-COG): a double-blind, pla-
considerable interest (••) to readers. cebo controlled trial. Lancet Neurol. 2008;7:683–689.
1. Wimo A, Guerchet M, Ali G-C, et al. The worldwide costs of demen- • One of very few clinical trials in older patients.
tia 2015 and comparisons with 2010. Alzheimers Dement. 21. Forette F, Seux M-L, Staessen JA, et al. The prevention of dementia
2017;13:1–7. with antihypertensive treatment: new evidence from the Systolic
10 T. LEBOUVIER ET AL.

Hypertension in Europe (Syst-Eur) study. Arch Intern Med. • A mice model showing the ACE inhibition is not deleterious,
2002;162:2046–2052. and could even decrease neurodegeneration.
• The first of clinical trials showing a marginally but significant 40. Hou D-R, Wang Y, Zhou L, et al. Altered angiotensin-converting
decrease in the incidence of dementia. enzyme and its effects on the brain in a rat model of Alzheimer
22. SPRINT MIND Investigators for the SPRINT Research Group, disease. Chin Med J. 2008;121:2320–2323.
Williamson JD, Pajewski NM, Auchus AP, et al., Effect of intensive 41. Asraf K, Torika N, Apte RN, et al. Microglial activation is modulated
vs standard blood pressure control on probable dementia: by captopril: in vitro and in vivo studies. Front Cell Neurosci.
a randomized clinical trial. JAMA. 2019;321:553–561. 2018;12:116.
23. SPRINT MIND Investigators for the SPRINT Research Group, 42. Kehoe PG, Davies NM, Martin RM, et al. Associations of angiotensin
Nasrallah IM, Pajewski NM, Auchus AP et al., Association of inten- targeting antihypertensive drugs with mortality and hospitalization
sive vs standard blood pressure control with cerebral white matter in primary care patients with dementia. J Alzheimers Dis.
lesions. JAMA. 2019;322:524–534. 2013;33:999–1008.
• This substudy showed a smaller increase in white matter lesion 43. Rist PM, Chalmers J, Arima H, et al. Baseline cognitive function,
volume by an intensive BP control. recurrent stroke, and risk of dementia in patients with stroke.
24. Rozzini L, Vicini Chilovi B, Bellelli G, et al. Effects of cholinesterase Stroke. 2013;44:1790–1795.
inhibitors appear greater in patients on established antihyperten- 44. Sink KM, Leng X, Williamson J, et al. Angiotensin-converting
sive therapy. Int J Geriat Psychiatry. 2005;20:547–551. enzyme inhibitors and cognitive decline in older adults with hyper-
25. Duron E, Rigaud A-S, Dubail D, et al. Effects of antihypertensive tension: results from the cardiovascular health study. Arch Intern
therapy on cognitive decline in Alzheimer’s disease. Am Med. 2009;169:1195–1202.
J Hypertens. 2009;22:1020–1024. 45. Gao Y, O’Caoimh R, Healy L, et al. Effects of centrally acting ACE
26. Bellew KM, Pigeon JG, Stang PE, et al. Hypertension and the rate of inhibitors on the rate of cognitive decline in dementia. BMJ Open.
cognitive decline in patients with dementia of the Alzheimer type. 2013;3:e002881.
Alzheimer Dis Assoc Disord. 2004;18:208–213. 46. Wharton W, Stein JH, Korcarz C, et al. The effects of ramipril in
27. Iadecola C, Yaffe K, Biller J, et al. Impact of Hypertension on individuals at risk for Alzheimer’s disease: results of a pilot clinical
cognitive function: a scientific statement from the American heart trial. J Alzheimers Dis. 2012;32:147–156.
association. Hypertension. 2016;68:1–56. 47. de Oliveira FF, Chen ES, Smith MC, et al. Pharmacogenetics of angio-
•• A comprehensive review on impact of hypertension on brain, tensin-converting enzyme inhibitors in patients with Alzheimer’s dis-
including at molecular level. ease dementia. Curr Alzheimer Res. 2018;15:386–398.
28. Gorelick PB, Scuteri A, Black SE, et al. Vascular contributions to 48. Nalivaeva NN, Turner AJ. Targeting amyloid clearance in
cognitive impairment and dementia: a statement for healthcare Alzheimer’s disease as a therapeutic strategy. Br J Pharmacol.
professionals from the American Heart Association/American 2019;4:190.
Stroke Association. Stroke. 2011;42:2672–2713. • This review focused on all the amyloid-degrading enzymes,
29. Sweeney MD, Montagne A, Sagare AP, et al. Vascular dysfunction- including angiotensin converting enzymes.
The disregarded partner of Alzheimer’s disease. Alzheimer’s 49. Liu J, Liu S, Tanabe C, et al. Differential effects of angiotensin II
Dementia. 2019;15:158–167. receptor blockers on Aβ generation. Neurosci Lett. 2014;567:51–56.
30. Petrovitch H, White LR, Izmirilian G, et al. Midlife blood pressure 50. Liu X, Wang Z, Xia Y, et al. Losartan-induced hypotension leads to
and neuritic plaques, neurofibrillary tangles, and brain weight at tau hyperphosphorylation and memory deficit. J Alzheimers Dis.
death: the HAAS. Honolulu-Asia aging study. NBA. 2000;21:57–62. 2014;40:419–427.
31. Cifuentes D, Poittevin M, Dere E, et al. Hypertension accelerates the 51. Zhao W, Wang J, Ho L, et al. Identification of antihypertensive
progression of Alzheimer-like pathology in a mouse model of the drugs which inhibit amyloid-beta protein oligomerization.
disease. Hypertension. 2015;65:218–224. J Alzheimers Dis. 2009;16:49–57.
32. Ashby EL, Miners JS, Kehoe PG, et al. Effects of hypertension and 52. Wang J, Ho L, Chen L, et al. Valsartan lowers brain beta-amyloid
anti-hypertensive treatment on Amyloid-β (Aβ) plaque load and protein levels and improves spatial learning in a mouse model of
Aβ-synthesizing and Aβ-degrading enzymes in frontal cortex. Alzheimer disease. J Clin Invest. 2007;117:3393–3402.
J Alzheimers Dis. 2016;50:1191–1203. 53. Mogi M, Li J-M, Tsukuda K, et al. Telmisartan prevented cognitive
33. Shih Y-H, Wu S-Y, Yu M, et al. Hypertension accelerates Alzheimer’s decline partly due to PPAR-gamma activation. Biochem Biophys
disease-related pathologies in Pigs and 3xTg Mice. Front Aging Res Commun. 2008;375:446–449.
Neurosci. 2018;10:73. 54. Tsukuda K, Mogi M, Iwanami J, et al. Cognitive deficit in
34. Zaman MA, Oparil S, Calhoun DA. Drugs targeting the amyloid-beta-injected mice was improved by pretreatment with
renin-angiotensin-aldosterone system. Nat Rev Drug Discovery. a low dose of telmisartan partly because of peroxisome
2002;1:621–636. proliferator-activated receptor-gamma activation. Hypertension.
35. Hemming ML, Selkoe DJ. Amyloid beta-protein is degraded by 2009;54:782–787.
cellular angiotensin-converting enzyme (ACE) and elevated by an • Positive effect of a ARB on cognition though the endothelial
ACE inhibitor. J Biol Chem. 2005;280:37644–37650. function.
36. Jalkute CB, Barage SH, Dhanavade MJ, et al. Molecular dynamics 55. Zhai Y, Yamashita T, Kurata T, et al. Strong reduction of low-density
simulation and molecular docking studies of Angiotensin convert- lipoprotein receptor/apolipoprotein E expressions by telmisartan in
ing enzyme with inhibitor lisinopril and amyloid Beta Peptide. cerebral cortex and hippocampus of stroke resistant spontaneously
Protein J. 2013;32:356–364. hypertensive rats. J Stroke Cerebrovasc Dis. 2014;23:2350–2361.
37. Bernstein KE, Koronyo Y, Salumbides BC, et al. Angiotensin- 56. Apolipoprotein BG. E and its receptors in Alzheimer’s disease: path-
converting enzyme overexpression in myelomonocytes prevents ways, pathogenesis and therapy. Nat Rev Neurosci. 2009;10:333–344.
Alzheimer’s-like cognitive decline. J Clin Invest. 2014;124:10 57. Shindo T, Takasaki K, Uchida K, et al. Ameliorative effects of telmisar-
00–1012. tan on the inflammatory response and impaired spatial memory in
38. Hemming ML, Selkoe DJ, Farris W. Effects of prolonged a rat model of Alzheimer’s disease incorporating additional cerebro-
angiotensin-converting enzyme inhibitor treatment on amyloid vascular disease factors. Biol Pharm Bull. 2012;35:2141–2147.
beta-protein metabolism in mouse models of Alzheimer disease. 58. Torika N, Asraf K, Danon A, et al. Telmisartan modulates glial
Neurobiol Dis. 2007;26:273–281. activation: in vitro and in vivo studies. PLoS One. 2016;11:
39. AbdAlla S, Langer A, Fu X, et al. ACE inhibition with captopril e0155823.
retards the development of signs of neurodegeneration in an 59. Tian M, Zhu D, Xie W, et al. Central angiotensin II-induced
animal model of Alzheimer’s disease. Int J Mol Sci. Alzheimer-like tau phosphorylation in normal rat brains. FEBS
2013;14:16917–16942. Lett. 2012;586:3737–3745.
EXPERT REVIEW OF NEUROTHERAPEUTICS 11

60. Takeda S, Sato N, Takeuchi D, et al. Angiotensin receptor blocker pre- 80. Yagami T, Ueda K, Asakura K, et al. Effects of endothelin B receptor
vented beta-amyloid-induced cognitive impairment associated with agonists on amyloid beta protein (25-35)-induced neuronal cell
recovery of neurovascular coupling. Hypertension. 2009;54:1345–1352. death. Brain Res. 2002;948:72–81.
61. Ongali B, Nicolakakis N, Tong X-K, et al. Angiotensin II type 1 81. Fu H, Li W, Lao Y, et al. Bis(7)-tacrine attenuates beta
receptor blocker losartan prevents and rescues cerebrovascular, amyloid-induced neuronal apoptosis by regulating L-type calcium
neuropathological and cognitive deficits in an Alzheimer’s disease channels. J Neurochem. 2006;98:1400–1410.
model. Neurobiol Dis. 2014;68:126–136. 82. Pierrot N, Ghisdal P, Caumont A-S, et al. Intraneuronal
• Interesting preclinical work about the positive effects of ARB amyloid-beta1-42 production triggered by sustained increase of
on cognition through decreasing oxidative stress in microves- cytosolic calcium concentration induces neuronal death.
sels without changes in blood pressure. J Neurochem. 2004;88:1140–1150.
62. Chiu W-C, Ho W-C, Lin M-H, et al. Angiotensin receptor blockers 83. Facchinetti F, Fasolato C, Del Giudice E, et al. Nimodipine selec-
reduce the risk of dementia. J Hypertens. 2014;32:938–947. tively stimulates beta-amyloid 1-42 secretion by a mechanism inde-
63. Hsu C-Y, Huang -C-C, Chan W-L, et al. Angiotensin-receptor block- pendent of calcium influx blockage. NBA. 2006;27:218–227.
ers and risk of Alzheimer’s disease in hypertension population–a 84. Paris D, Ait-Ghezala G, Bachmeier C, et al. The spleen tyrosine
nationwide cohort study. Circ J. 2013;77:405–410. kinase (Syk) regulates Alzheimer amyloid-β production and Tau
64. Skoog I, Lithell H, Hansson L, et al. Effect of baseline cognitive hyperphosphorylation. J Biol Chem. 2014;289:33927–33944.
function and antihypertensive treatment on cognitive and cardio- • This paper provides a deeper understanding of the inhibition
vascular outcomes: study on COgnition and Prognosis in the of amyloidogenesis of CCBs.
Elderly (SCOPE. Am J Hypertens. 2005;18:1052–1059. 85. Bachmeier C, Beaulieu-Abdelahad D, Mullan M, et al. Selective
65. Zanchetti A, Elmfeldt D. Findings and implications of the study on dihydropyiridine compounds facilitate the clearance of β-amyloid
cognition and prognosis in the elderly (SCOPE) - a review. Blood across the blood-brain barrier. Eur J Pharmacol. 2011;659:124–129.
Press. 2006;15:71–79. 86. Paris D, Quadros A, Humphrey J, et al. Nilvadipine antagonizes both
66. Saxby BK, Harrington F, Wesnes KA, et al. Candesartan and cogni- Abeta vasoactivity in isolated arteries, and the reduced cerebral
tive decline in older patients with hypertension: a substudy of the blood flow in APPsw transgenic mice. Brain Res. 2004;999:53–61.
SCOPE trial. Neurology. 2008;70:1858–1866. 87. Ekinci FJ, Ortiz D, Shea TB. Okadaic acid mediates tau phosphoryla-
67. Wharton W, Goldstein FC, Tansey MG, et al. Rationale and design of tion via sustained activation of the L-voltage-sensitive calcium
the mechanistic potential of antihypertensives in preclinical channel. Brain Res Mol Brain Res. 2003;117:145–151.
Alzheimer’s (HEART) trial. JAD. 2017;61:815–824. 88. Sanz JM, Chiozzi P, Colaianna M, et al. Nimodipine inhibits IL-1β
• Interesting rationale and design of an on-going clinical trial. release stimulated by amyloid β from microglia. Br J Pharmacol.
68. Szabo-Reed AN, Vidoni E, Binder EF, et al. Rationale and methods 2012;167:1702–1711.
for a multicenter clinical trial assessing exercise and intensive 89. Gaasch JA, Geldenhuys WJ, Lockman PR, et al. Voltage-gated cal-
vascular risk reduction in preventing dementia (rrAD Study). cium channels provide an alternate route for iron uptake in neu-
Contemp Clin Trials. 2019;79:44–54. ronal cell cultures. Neurochem Res. 2007;32:1686–1693.
69. Heneka MT, Landreth GE, Hüll M. Drug insight: effects mediated by 90. Bostanci MÖ, Bagirici F. Blocking of L-type calcium channels pro-
peroxisome proliferator-activated receptor-gamma in CNS tects hippocampal and nigral neurons against iron neurotoxicity.
disorders. Nat Clin Pract Neurol. 2007;3:496–504. The role of L-type calcium channels in iron-induced neurotoxicity.
70. Michel MC, Foster C, Brunner HR, et al. A systematic comparison of Int J Neurosci. 2013;123:876–882.
the properties of clinically used angiotensin II type 1 receptor 91. López-Arrieta JM, Birks J. Nimodipine for primary degenerative,
antagonists. Pharmacol Rev. 2013;65:809–848. mixed and vascular dementia. Cochrane Database Syst Rev.
71. Kume K, Hanyu H, Sakurai H, et al. Effects of telmisartan on cogni- 2002;14:CD000147.
tion and regional cerebral blood flow in hypertensive patients with 92. Caramelli P, Laks J, Palmini ALF, et al. Effects of galantamine and
Alzheimer’s disease. Geriatr Gerontol Int. 2012;12:207–214. galantamine combined with nimodipine on cognitive speed and
• Evidence that showed ARB could address vascular dysfunction. quality of life in mixed dementia: a 24-week, randomized,
72. Ozawa Y, Hayashi K, Kobori H. New generation calcium channel placebo-controlled exploratory trial (the REMIX study. Arq
blockers in hypertensive treatment. Curr Hypertens Rev. Neuropsiquiatr. 2014;72:411–417.
2006;2:103–111. 93. Matsuda H, Araki N, Kuji I, et al. Effect of nilvadipine on regional
73. Zamponi GW. Targeting voltage-gated calcium channels in neuro- cerebral blood flow in a patient with early Alzheimer disease. Clin
logical and psychiatric diseases. Nat Rev Drug Discov. Nucl Med. 2008;33:34–35.
2016;15:19–34. 94. Kennelly SP, Abdullah L, Paris D, et al. Demonstration of safety in
74. Weiss JH, Pike CJ, Cotman CW. Ca2+ channel blockers attenuate Alzheimer’s patients for intervention with an anti-hypertensive
beta-amyloid peptide toxicity to cortical neurons in culture. drug Nilvadipine: results from a 6-week open label study.
J Neurochem. 1994;62:372–375. Int J Geriat Psychiatry. 2011;26:1038–1045.
75. Freir DB, Costello DA, Herron CE. A beta 25-35-induced depression 95. Kennelly S, Abdullah L, Kenny RA, et al. Apolipoprotein E genotype-
of long-term potentiation in area CA1 in vivo and in vitro is specific short-term cognitive benefits of treatment with the anti-
attenuated by verapamil. J Neurophysiol. 2003;89:3061–3069. hypertensive nilvadipine in Alzheimer’s patients–an open-label
76. Iwasaki K, Egashira N, Takagaki Y, et al. Nilvadipine prevents the trial. Int J Geriat Psychiatry. 2012;27:415–422.
impairment of spatial memory induced by cerebral ischemia com- 96. Lawlor B, Kennelly S, O’Dwyer S, et al. NILVAD protocol:
bined with beta-amyloid in rats. Biol Pharm Bull. 2007;30:698–701. a European multicentre double-blind placebo-controlled trial of
77. Gholamipour-Badie H, Naderi N, Khodagholi F, et al. L-type calcium nilvadipine in mild-to-moderate Alzheimer’s disease. BMJ Open.
channel blockade alleviates molecular and reversal spatial learning 2014;4:e006364.
and memory alterations induced by entorhinal amyloid pathology 97. Lawlor B, Segurado R, Kennelly S, et al. Nilvadipine in mild to
in rats. Behav Brain Res. 2013;237:190–199. moderate Alzheimer disease: A randomised controlled trial. PLoS
78. Paris D, Bachmeier C, Patel N, et al. Selective antihypertensive Med. 2018;15:e1002660.
dihydropyridines lower Aβ accumulation by targeting both the •• This recent clinical trial testing Nilvadipine for patients with
production and the clearance of Aβ across the blood-brain barrier. mild to moderate AD failed to demonstrate any effect on
Mol Med. 2011;17:149–162. cognitive outcome, but there was less decline in some prede-
79. Ho R, Ortiz D, Shea TB. Amyloid-beta promotes calcium influx and fined subgroups.
neurodegeneration via stimulation of L voltage-sensitive calcium 98. de Jong DLK, de Heus RAA, Rijpma A, et al. Effects of nilvadipine on
channels rather than NMDA channels in cultured neurons. cerebral blood flow in patients with Alzheimer Disease.
J Alzheimers Dis. 2001;3:479–483. Hypertension. 2019;74:413–420.
12 T. LEBOUVIER ET AL.

99. Dobarro M, Gerenu G, Ramírez MJ. Propranolol reduces cognitive 119. O’Donnell MJ, Xavier D, Liu L, et al. Risk factors for ischaemic and
deficits, amyloid and tau pathology in Alzheimer’s transgenic mice. intracerebral haemorrhagic stroke in 22 countries (the INTERSTROKE
Int J Neuropsychopharmacol. 2013;16:2245–2257. study): a case-control study. Lancet. 2010;376:112–123.
100. Dobarro M, Orejana L, Aguirre N, et al. Propranolol restores cogni- 120. Dufouil C, Chalmers J, Coskun O, et al. Effects of blood pressure
tive deficits and improves amyloid and Tau pathologies in a lowering on cerebral white matter hyperintensities in patients with
senescence-accelerated mouse model. Neuropharmacology. stroke: the PROGRESS (Perindopril protection against recurrent
2013;64:137–144. stroke study) Magnetic resonance imaging substudy. Circulation.
101. Dobarro M, Orejana L, Aguirre N, et al. Propranolol reduces cogni- 2005;112:1644–1650.
tive deficits, amyloid β levels, tau phosphorylation and insulin 121. Friedman JI, Tang CY, de Haas HJ, et al. Brain imaging changes
resistance in response to chronic corticosterone administration. associated with risk factors for cardiovascular and cerebrovascular
Int J Neuropsychopharmacol. 2013;16:1351–1360. disease in asymptomatic patients. JACC Cardiovasc Imaging.
102. Wang J, Wright HM, Vempati P, et al. Investigation of nebivolol as 2014;7:1039–1053.
a novel therapeutic agent for the treatment of Alzheimer’s disease. 122. Toledo J, Arnold S, Raible K, et al. Contribution of cerebrovascular
J Alzheimers Dis. 2013;33:1147–1156. disease in autopsy confirmed neurodegenerative disease cases in the
103. Saber AJ, Cain DP. Combined beta-adrenergic and cholinergic antagon- National Alzheimer’s coordinating centre. Brain. 2013;136:2697–2706.
ism produces behavioral and cognitive impairments in the water maze: 123. Jellinger KA. The enigma of mixed dementia. Alzheimer’s
implications for Alzheimer disease and pharmacotherapy with Dementia. 2007;3:40–53.
beta-adrenergic antagonists. Neuropsychopharmacology. 2003;28:12 124. Zlokovic BV. Neurovascular pathways to neurodegeneration in
47–1256. Alzheimer’s disease and other disorders. Nat Rev Neurosci.
104. Kenton L, Boon F, Cain DP. Combined but not individual adminis- 2011;12:723–738.
tration of beta-adrenergic and serotonergic antagonists impairs 125. Regan C, Katona C, Walker Z, et al. Relationship of vascular risk to the
water maze acquisition in the rat. Neuropsychopharmacology. progression of Alzheimer disease. Neurology. 2006;67:1357–1362.
2008;33:1298–1311. 126. Ezzati A, Wang C, Lipton RB, et al. Association between vascular
105. Gelber RP, Ross GW, Petrovitch H, et al. Antihypertensive medica- pathology and rate of cognitive decline independent of Alzheimer’s
tion use and risk of cognitive impairment: the Honolulu-Asia Aging disease pathology. J Am Geriatr Soc. 2017;65:1836–1841.
Study. Neurology. 2013;81:888–895. 127. Korf ESC, White LR, Scheltens P, et al. Midlife blood pressure and
106. Yasar S, Xia J, Yao W, et al. Antihypertensive drugs decrease risk of the risk of hippocampal atrophy: the Honolulu Asia Aging Study.
Alzheimer disease: Ginkgo evaluation of memory study. Neurology. Hypertension. 2004;44:29–34.
2013;81:896–903. 128. Hoffman LB, Schmeidler J, Lesser GT, et al. Less Alzheimer disease
107. Ruilope LM, Dukat A, Böhm M, et al. Blood-pressure reduction with neuropathology in medicated hypertensive than nonhypertensive
LCZ696, a novel dual-acting inhibitor of the angiotensin II receptor persons. Neurology. 2009;72:1720–1726.
and neprilysin: a randomised, double-blind, placebo-controlled, 129. Thal DR, Griffin WST, Vos RAI, et al. Cerebral amyloid angiopathy and its
active comparator study. Lancet. 2010;375:1255–1266. relationship to Alzheimer’s disease. Acta Neuropathol. 2008;115:599–609.
108. McMurray JJV, Packer M, Desai AS, et al. Angiotensin-neprilysin 130. Charidimou A, Gang Q, Werring DJ. Sporadic cerebral amyloid
inhibition versus enalapril in heart failure. N Engl J Med. angiopathy revisited: recent insights into pathophysiology and
2014;371:993–1004. clinical spectrum. J Neurol Neurosurg Psychiatry. 2012;83:124–137.
109. Vodovar N, Paquet C, Mebazaa A, et al. Neprilysin, cardiovascular, 131. Hughes TM, Kuller LH, Barinas-Mitchell EJM, et al. Arterial stiffness
and Alzheimer’s diseases: the therapeutic split? Eur Heart J. and β-amyloid progression in nondemented elderly adults. JAMA
2015;36:902–905. Neurol. 2014;71:562–568.
110. Iwata N, Tsubuki S, Takaki Y, et al. Metabolic regulation of brain • This paper provides some rationales about how hypertension
Abeta by neprilysin. Science. 2001;292:1550–1552. could impair the perivascular drainage.
111. Hüttenrauch M, Baches S, Gerth J, et al. Neprilysin deficiency alters the 132. Sweeney MD, Sagare AP, Zlokovic BV. Blood-brain barrier break-
neuropathological and behavioral phenotype in the 5XFAD mouse down in Alzheimer disease and other neurodegenerative disorders.
model of Alzheimer’s disease. J Alzheimers Dis. 2015;44:1291–1302. Nat Rev Neurol. 2018;14:133–150.
112. Campbell DJ. Long-term neprilysin inhibition - implications for • A comprehensive review focused on the importance of blood–
ARNIs. Nat Rev Cardiol. 2017;14:171–186. brain barrier.
113. Schoenfeld HA, West T, Verghese PB, et al. The effect of angiotensin 133. Ozacmak VH, Sayan H, Cetin A, et al. AT1 receptor blocker
receptor neprilysin inhibitor, sacubitril/valsartan, on central ner- candesartan-induced attenuation of brain injury of rats subjected to
vous system amyloid-β concentrations and clearance in the cyno- chronic cerebral hypoperfusion. Neurochem Res. 2007;32:1314–1321.
molgus monkey. Toxicol Appl Pharmacol. 2017;323:53–65. 134. Fleegal-DeMotta MA, Doghu S, Banks WA. Angiotensin II modulates
114. Langenickel TH, Tsubouchi C, Ayalasomayajula S, et al. The effect of BBB permeability via activation of the AT(1) receptor in brain
LCZ696 (sacubitril/valsartan) on amyloid-β concentrations in cere- endothelial cells. J Cereb Blood Flow Metab. 2009;29:640–647.
brospinal fluid in healthy subjects. Br J Clin Pharmacol. 135. Pelisch N, Hosomi N, Ueno M, et al. Blockade of AT1 receptors
2016;81:878–890. protects the blood-brain barrier and improves cognition in Dahl
115. Katsouri L, Vizcaychipi MP, McArthur S, et al. Prazosin, an α(1)- salt-sensitive hypertensive rats. Am J Hypertens. 2011;24:362–368.
adrenoceptor antagonist, prevents memory deterioration in the 136. Min L-J, Mogi M, Shudou M, et al. Peroxisome proliferator-activated
APP23 transgenic mouse model of Alzheimer’s disease. Neurobiol receptor-γ activation with angiotensin II type 1 receptor blockade is
Aging. 2013;34:1105–1115. pivotal for the prevention of blood-brain barrier impairment and cogni-
116. Wang LY, Shofer JB, Rohde K, et al. Prazosin for the treatment of tive decline in type 2 diabetic mice. Hypertension. 2012;59:1079–1088.
behavioral symptoms in patients with Alzheimer disease with agitation 137. Appleby BS, Nacopoulos D, Milano N, et al. A review: treatment of
and aggression. Am J Geriatr Psychiatry. 2009;17:744–751. Alzheimer’s disease discovered in repurposed agents. Dement
117. Arya U, Dwivedi H, Subramaniam JR. Reserpine ameliorates Abeta Geriatr Cognit Disord. 2013;35:1–22.
toxicity in the Alzheimer’s disease model in Caenorhabditis ele- 138. Barone E, Di Domenico F, Butterfield DA. Statins more than cholesterol
gans. Exp Gerontol. 2009;44:462–466. lowering agents in Alzheimer disease: their pleiotropic functions as
118. Saharia K, Arya U, Kumar R, et al. Reserpine modulates neurotransmitter potential therapeutic targets. Biochem Pharmacol. 2014;88:605–616.
release to extend lifespan and alleviate age-dependent Aβ proteotoxi- 139. Corbett A, Pickett J, Burns A, et al. Drug repositioning for
city in Caenorhabditis elegans. Exp Gerontol. 2012;47:188–197. Alzheimer’s disease. Nat Rev Drug Discovery. 2012;11:833–846.
EXPERT REVIEW OF NEUROTHERAPEUTICS 13

140. Corbett A, Williams G, Ballard C. Drug repositioning: an opportunity to 146. Qiu C, Winblad B, Fratiglioni L. Low diastolic pressure and risk of
develop novel treatments for Alzheimer’s disease. Pharmaceuticals (Basel). dementia in very old people: a longitudinal study. Dement Geriatr
2013;6:1304–1321. Cognit Disord. 2009;28:213–219.
141. Dubois B, Feldman HH, Jacova C, et al. Advancing research diagnostic 147. De Heus R, Donders RSantoso A, et al. Blood pressure lowering with
criteria for Alzheimer’s disease: the IWG-2 criteria. Lancet Neurol. nilvadipine in patients with mild-to-moderate alzheimer disease
2014;13:614–629. does not increase the prevalence of orthostatic hypotension. J
142. Jack CR, Bennett DA, Blennow K, et al. NIA-AA Research Framework: Am Heart Assoc. 2019;8:e011938.
toward a biological definition of Alzheimer’s disease. Alzheimers 148. Barthold D, Joyce G, Wharton W, et al. The association of multiple
Dement. 2018;14:535–562. anti-hypertensive medication classes with Alzheimer’s disease inci-
143. Banerjee G, Wilson D, Jäger H, et al. Novel imaging techniques in dence across sex, race, and ethnicity. PLoS One. 2018;13:e0206705.
cerebral small vessel diseases and vascular cognitive impairment. •• Interesting study which support the need toward a more per-
Biochim Biophys Acta. 2016;1862:926–938. sonalized therapeutic strategy.
144. Villemagne VL, Doré V, Burnham SC, et al. Imaging tau and amy- 149. Roshandel G, Khoshnia M, Poustchi H, et al. Effectiveness of polypill for
loid-β proteinopathies in Alzheimer disease and other conditions. primary and secondary prevention of cardiovascular diseases (PolyIran):
Nat Rev Neurol. 2018;14:225–236. a pragmatic, cluster-randomised trial. Lancet. 2019;394:672–683.
145. Qiu C, Strauss von E, Winblad B, et al. Decline in blood pressure 150. Valenti R, Pantoni L, Markus HS. Treatment of vascular risk factors in
over time and risk of dementia: a longitudinal study from the patients with a diagnosis of Alzheimer’s disease: a systematic
Kungsholmen project. Stroke. 2004;35:1810–1815. review. BMC Med. 2014;12:160.

You might also like