Cilostazol A
Cilostazol A
Cilostazol A
Molecular Sciences
Review
The Role of Cilostazol, a Phosphodiesterase-3 Inhibitor, in the
Development of Atherosclerosis and Vascular Biology: A Review
with Meta-Analysis
Minji Sohn and Soo Lim *
Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University
College of Medicine, Seongnam 13620, Republic of Korea; [email protected]
* Correspondence: [email protected]
Abstract: Atherosclerotic cardiovascular disease (ASCVD) stands as the leading global cause of
mortality. Addressing this vital and pervasive condition requires a multifaceted approach, in which
antiplatelet intervention plays a pivotal role, together with antihypertensive, antidiabetic, and lipid-
lowering therapies. Among the antiplatelet agents available currently, cilostazol, a phosphodiesterase-
3 inhibitor, offers a spectrum of pharmacological effects. These encompass vasodilation, the impedi-
ment of platelet activation and aggregation, thrombosis inhibition, limb blood flow augmentation,
lipid profile enhancement through triglyceride reduction and high-density lipoprotein cholesterol
elevation, and the suppression of vascular smooth muscle cell proliferation. However, the role of
cilostazol has not been clearly documented in many guidelines for ASCVD. We comprehensively
reviewed the cardiovascular effects of cilostazol within randomized clinical trials that compared it to
control or active agents and involved individuals with previous coronary artery disease or stroke,
as well as those with no previous history of such conditions. Our approach demonstrated that the
administration of cilostazol effectively reduced adverse cardiovascular events, although there was
less evidence regarding its impact on myocardial infarction. Most studies have consistently reported
its favorable effects in reducing intermittent claudication and enhancing ambulatory capacity in
patients with peripheral arterial disease. Furthermore, cilostazol has shown promise in mitigating
restenosis following coronary stent implantation in patients with acute coronary syndrome. While
Citation: Sohn, M.; Lim, S. The Role research from more diverse regions is still needed, our findings shed light on the broader implications
of Cilostazol, a Phosphodiesterase-3 of cilostazol in the context of atherosclerosis and vascular biology, particularly for individuals at high
Inhibitor, in the Development of risk of ASCVD.
Atherosclerosis and Vascular Biology:
A Review with Meta-Analysis. Int. J. Keywords: atherosclerosis; cardiovascular diseases; cilostazol; platelet aggregation inhibitors; aspirin
Mol. Sci. 2024, 25, 2593. https://
doi.org/10.3390/ijms25052593
These vascular diseases are mainly caused by atherosclerosis, which starts with the
development of fibrofatty lesions known as atherosclerotic plaques. Several mechanisms
are involved in this process: the accumulation of modified low-density lipoprotein (LDL)
in the intima, the attraction of pro-inflammatory monocytes and T lymphocytes to the
arterial wall, the proliferation and migration of vascular smooth muscle cells (VSMCs), the
aggravation of oxidative stress and the increase in adhesion molecules, and the aggregation
of platelets [1]. The rupture of the fibrous cap in an atherosclerotic plaque can result in the
development of a life-threatening ASCVD, such as myocardial infarction and stroke, both of
which can also lead to various complications that significantly impact the patient’s quality of
life [6,7]. Furthermore, ASCVD is commonly asymptomatic up until the occurrence of acute
coronary syndrome or stroke [8]. Therefore, preventive measures and early interventions
aimed at reducing the risk of these conditions are crucial.
Type 2 diabetes mellitus (T2DM) is closely linked to an increased risk of CAD and cere-
brovascular disease. Patients with T2DM have a 2–4-fold higher CVD risk compared to that
of the general population, regardless of the presence/absence of other risk factors [9]. Thus,
diabetes mellitus is regarded as being equivalent to the risk of CAD. Furthermore, ASCVD
is the principal cause of death and disability among patients with diabetes mellitus [10].
Diabetes mellitus is characterized by insulin resistance, dyslipidemia, inflammation, and
hypercoagulability, which exacerbate the underlying causes of atherosclerosis and heart
failure [11]. To date, the cardiovascular benefit of multifactorial risk-reduction approaches
using antidiabetic agents, antihypertensive drugs, and lipid-lowering therapy has been
proven, thus reducing the incidence of cardiovascular complications [12]. However, the cur-
rent therapeutic strategies are not sufficient for preventing adverse cardiovascular events,
and a substantial number of patients with diabetes mellitus still face a risk of CVD, leading
to the term “residual risk” [12].
For the management of the residual risk, international guidelines from the USA
and Europe recommend the use of antiplatelet agents as a main management strategy
in patients with diabetes and a high CVD risk [13,14]. Hence, antiplatelet agents such
as aspirin, clopidogrel, and cilostazol could be contemplated in the management of the
residual risk, pending additional supportive evidence.
Cilostazol is an antiplatelet agent that inhibits phosphodiesterase-3 (PDE-3) and in-
creases cyclic adenosine monophosphate (cAMP) concentrations, leading to the inhibition
of platelet aggregation and thrombus formation [15]. PDE-3 is expressed in cell populations
such as platelets, VSMCs, cardiac myocytes, and adipocytes [16]. Therefore, cilostazol is
expected to have some effects in these tissues.
Cilostazol has been approved for improving intermittent claudication symptoms
in various countries. In further research, cilostazol treatment reduced the progression of
atherosclerosis in patients with carotid artery stenosis [17,18] and the recurrence of stroke in
patients with cerebrovascular disease [19,20]. In addition, previous studies have shown that
cilostazol therapy is beneficial for CAD when administered immediately after percutaneous
coronary intervention or in addition to other antiplatelet agents [21,22].
In 2015, a critical review of the literature regarding cilostazol and its role against
CVD was reported [23]. Since then, many basic and clinical studies have been published;
however, its role has not been reviewed systematically. Moreover, cilostazol has not been
well documented in the literature with regard to its role in CVD and diabetes mellitus. In
this review, we discuss the mechanism of action of cilostazol and evaluate the efficacy and
safety of the drug based on the results of clinical trials. Furthermore, we comprehensively
explore the role of cilostazol in the prevention and treatment of atherosclerosis.
aspirin for CVD in patients with T2DM reported an absence of significant differences in the
incidence of CVD between the aspirin treatment group and the non-aspirin group [25,26]. A
meta-analysis of several clinical studies in which aspirin was administered for the primary
prevention of CVD also showed that aspirin did not reduce the incidence of CVD in patients
with diabetes [27]. The six-year follow-up of the primary preventive effect of aspirin against
CVD in more than 14,000 Japanese elderly patients also failed to demonstrate the beneficial
effect of aspirin on the incidence of CVD [28].
Moreover, it was reported that the use of aspirin not only increased the number of
adverse events, such as major bleeding and gastrointestinal discomfort, but also did not
reduce the risk of cardiovascular events in primary prevention (Table 1) [25,26,28–34].
There is some evidence from basic studies suggesting that aspirin might not be suffi-
cient to reduce abnormal platelet activation. The platelet multidrug resistance protein 4
(MRP4) is upregulated after aspirin administration, which could represent one of the
mechanisms underlying the high residual platelet reactivity after aspirin intake [35]. Impor-
tantly, patients with persistent high platelet reactivity while on aspirin therapy exhibited
an increased risk of recurrent cardiovascular events [36].
Based on these findings, the role of aspirin in this context has changed drastically
since 2018. The US Preventive Services Task Force did not recommend the initiation of low-
dose aspirin among patients aged 60 years or older [37]. The American Heart Association
(AHA) and American College of Cardiology (ACC) Foundation’s statement on aspirin
for the primary prevention of cardiovascular events in individuals with diabetes suggests
that low-dose aspirin should not be administered to those with a low risk for ASCVD
(10-year risk < 5%), but can be considered in patients with an increased cardiovascular risk
(10-year risk > 10%) [38,39]. Unfortunately, no direct evidence from a clinical trial supports
these risk-based treatment recommendations, resulting in controversy in the primary
prevention among patients with a high CVD risk. Moreover, the primary preventive effect
of aspirin was not clearly demonstrated, even for high-risk patients, such as those with
T2DM (Table 1).
In contrast, the guidelines for the secondary prevention of diabetes recommend aspirin
therapy (75–162 mg daily) for individuals with diabetes and a history of ASCVD [13].
Nevertheless, concerns regarding the side effects of this therapy remain unaddressed.
In the Aspirin in Reducing Events in the Elderly (ASPREE) study, cancer-related death
occurred in 3.1% of the participants in the aspirin group and in 2.3% of those in the placebo
group (hazard ratio: 1.31; 95% confidence interval (CI): 1.10–1.56), indicating a higher
cancer mortality rate in the aspirin therapy compared with that of the placebo group [33].
Although the risk of cancer development was not confirmed in other studies (Table 1),
concerns regarding this issue remain.
Int. J. Mol. Sci. 2024, 25, 2593 4 of 26
Table 1. Randomized controlled studies with aspirin for preventing primary cardiovascular events since 2000.
Patient’s
Trial Information * Efficacy and Safety Outcomes
Bibliographic Characteristics
Information Patients’ Men (%)/Age Diabetes Cardiovascular
Population Duration ** Bleeding Risks Cancer Risks
Number (Years) Comorbidity Events ***
No difference in
RR of 3P-MACE a Severe bleedings more
Participants with at least cancer risks with
with aspirin vs. frequent in the aspirin
PPP study [29] one already known major 5 years 4495 42.5/64.4 16.5% aspirin vs.
no-aspirin: group (1.1% vs. 0.3%;
cardiovascular risk factor no-aspirin
0.71 (0.48–1.04) p < 0.0008)
(3.9% vs. 3.5%)
Female health Any gastrointestinal
RR of 3P-MACE a
professionals, aged ≥45 bleeding: RR
with aspirin vs.
WHS study [30] and without a history of mean 10.1 years 39,876 0/54.6 2.6% 1.22 (1.10–1.34) Not reported
placebo: 0.91
cardiovascular disease Requiring transfusion:
(0.80–1.03)
or cancer RR 1.40 (1.07–1.83)
No difference in
HR of atherosclerotic significant
Patients with events b with aspirin gastrointestinal
JPAD study [25] mean 4.37 years 2539 54.6/64 100% Not reported
type 2 diabetes vs. no-aspirin: bleeding with aspirin
0.80 (0.58–1.10) vs. no-aspirin
(1.0% vs. 0.3%)
Participants aged ≥40
with type 1 or type 2 No difference in No difference in
No difference in
diabetes and an ankle atherosclerotic events gastrointestinal
POPADAD median 6.7 years c with aspirin vs. malignancy with
brachial pressure index of 1276 44.1/60.3 100% bleeding with aspirin
study [26] (4.5~8.6 years) aspirin vs. placebo
0.99 or less but no no-aspirin: vs. no-aspirin:
(8.3% vs. 10.7%)
symptomatic 0.98 (0.76–1.26) 0.90 (0.53–1.52)
cardiovascular disease
Participants aged 60 to Major gastrointestinal
85 years, presenting with HR of 3P-MACE a and extracranial
hypertension, 6 years (median with aspirin vs. bleedings more
JPPP study [28] 14,658 42.3/70.5 33.9% Not reported
dyslipidemia, or diabetes 5.02 years) no-aspirin: frequent in the aspirin
mellitus without 0.94 (0.77–1.15) group (0.1% vs. 0.07%;
cardiovascular disease p < 0.001)
Int. J. Mol. Sci. 2024, 25, 2593 5 of 26
Table 1. Cont.
Patient’s
Trial Information * Efficacy and Safety Outcomes
Bibliographic Characteristics
Information Patients’ Men (%)/Age Diabetes Cardiovascular
Population Duration ** Bleeding Risks Cancer Risks
Number (Years) Comorbidity Events ***
RR of serious vascular
Patients with type 2 RR of any major
ASCEND at least 7 years events d with aspirin RR of any cancer:
diabetes without 15,480 62.6/63.3 100% bleeding:
study [31] (mean 7.4 years) vs. placebo: 1.01 (0.92–1.11)
cardiovascular disease 1.29 (1.09–1.52)
0.88 (0.79–0.97)
Participants aged
≥55 years (men) or
HR of 3P-MACE a No difference in
≥60 years (women) and HR of gastrointestinal
ARRIVE study 6 years (mean with aspirin vs. cancer with aspirin
had moderate 12,546 70.4/63.9 0% bleeding:
[32] 5 years) no-aspirin: vs. placebo (not
cardiovascular risk, 2.11 (1.39–3.28)
0.79 (0.61–1.02) sufficient events)
without at high risk of
bleeding or diabetes
HR of ischemic HR of major
Elderly participants who HR of cancer-related
events-related deaths hemorrhage-related
ASPREE did not have 6 years (median deaths with aspirin
19,114 43.6/median 74 10.8 with aspirin vs. deaths with aspirin vs.
study [33] cardiovascular disease, 4.7 years) vs. placebo:
placebo: placebo:
dementia, or disability 1.31 (1.10–1.56)
0.82 (0.62–1.08) 1.13 (0.66–1.94)
Participants without No difference in major
HR of 3P-MACE a by HR of cancer with
Yusuf S. et al., cardiovascular disease 6 years (mean bleeding with aspirin
5713 47.1/63.9 36.7% aspirin vs. placebo: aspirin vs. placebo:
2021 [34] who had an elevated 4.6 years) vs. placebo
0.86 (0.67–1.10) 0.83 (0.54–1.27)
INTERHEART Risk Score (1.5% vs. 1.3%)
Abbreviations: 3P-MACE, 3-point major adverse cardiovascular events; HR, hazard ratio; and RR, risk ratio. * Aspirin (100 mg) was compared to placebo or control therapy. ** Study
drug was administered during the whole study period. *** Definition of cardiovascular events were different between the groups: a 3P-MACE: cardiovascular death, non-fatal,
myocardial infarction, and stroke. b Composite of sudden death; death from coronary, cerebrovascular, and aortic causes; non-fatal acute myocardial infarction; unstable angina; newly
developed exertional angina; nonfatal ischemic and hemorrhagic stroke; transient ischemic attack; or nonfatal aortic and peripheral vascular disease (arteriosclerosis obliterans, aortic
dissection, mesenteric arterial thrombosis). c Death from coronary heart disease or stroke, non-fatal myocardial infarction or stroke, or above ankle amputation for critical limb ischemia.
d Composite of nonfatal myocardial infarction, nonfatal stroke (excluding confirmed intracranial hemorrhage) or transient ischemic attack, or death from any vascular cause (excluding
3. Beneficial RoleRole
3. Beneficial of Cilostazol in the
of Cilostazol in Development of Atherosclerosis
the Development of Atherosclerosis
3.1. 3.1.
Antiplatelet Activity
Antiplatelet Activity
Cilostazol inhibits
Cilostazol platelet
inhibits function
platelet by inhibiting
function PDE-3
by inhibiting and increasing
PDE-3 cAMPcAMP
and increasing levels lev-
[40]els
(Figure 1). Platelet
[40] (Figure reactivity
1). Platelet is mediated
reactivity by the
is mediated byintracellular calcium
the intracellular levels,
calcium which
levels, which
induce a molecular
induce a molecular cascade including
cascade cAMP,
including cAMP,resulting in platelet
resulting degranulation,
in platelet degranulation, confor-
confor-
mational
mational changes,
changes, and and aggregation
aggregation [41]. [41]. Elevated
Elevated cAMPcAMP levels levels upregulate
upregulate the activity
the activity of
of protein
protein kinase A kinase
(PKA),A which
(PKA),phosphorylates
which phosphorylates key intermediary
key intermediary moleculesmolecules that are
that are essen-
essential
tial for plateletforaggregation
platelet aggregation
[42]. PKA [42]. PKA
has also hastied
been alsotobeen tied to the downregulation
the downregulation of platelet of
platelet
adhesion adhesion
through the through the phosphorylation
phosphorylation of the GP-Ib-IXof the GP-Ib-IX
complex, thecomplex,
inhibitionthe
of inhibition
intracel- of
lularintracellular
calcium store calcium storethromboxane
release, release, thromboxane
receptor receptor desensitization,
desensitization, and theand the inhibition
inhibition of
of the phosphorylation
the phosphorylation of myosin
of myosin light kinase
light chain chain kinase
[43]. [43].
Recent evidence
Recent hashas
evidence suggested
suggesteda role forfor
a role cilostazol in in
cilostazol thethe
inhibition
inhibitionofof
MRP4
MRP4 [44]
[44]and
and in
in overcoming a high on-aspirin-treatment residual platelet reactivity in a
overcoming a high on-aspirin-treatment residual platelet reactivity in a cAMP-independent cAMP-inde-
pendent manner
manner [45]. MRP4,
[45]. MRP4, which which is expressed
is expressed in platelets
in platelets primarily
primarily in theinmembranes
the membranesof dense
of dense granules [46], promotes platelet aggregation by transporting
granules [46], promotes platelet aggregation by transporting cyclic nucleotides, cyclic nucleotides,
including
including
cAMP cAMP and cGMP,
and cGMP, outside outside of platelets
of platelets [47]. MRP4
[47]. MRP4 has also
has also beenbeen
shown shown to de-
to desensitize
sensitize platelets
platelets to thetoinhibitory
the inhibitory effects
effects of endothelial-derived
of endothelial-derived nitric
nitric oxide
oxide andand sensitize
sensitize them
them to ADP-mediated activation [44]. Thus, the inactivation of MRP4 and
to ADP-mediated activation [44]. Thus, the inactivation of MRP4 and the inhibition of the inhibition
of phosphodiesterase-4
phosphodiesterase-4by bycilostazol
cilostazolare
arereported
reported toto
have
have synergistic and
synergistic andpotent
potenttherapeu-
therapeutic
tic antiplatelet effects [46].
antiplatelet effects [46].
Int. J. Mol. Sci. 2024, 25, 2593 7 of 26
The clinical findings of decreased platelet reactivity after cilostazol treatment have
been described in various populations, including patients with acute coronary syndrome
and ischemic stroke [40,48], and even when administered in addition to aspirin and clopido-
grel [49]. Cilostazol is likely to be more effective in inhibiting platelet activation, especially
in patients with diabetes mellitus. Platelets themselves have insulin receptors which, when
activated, initiate a molecular cascade, leading to increased cAMP levels and platelet inhi-
bition [50]. Insulin resistance is associated with an attenuated P2Y1 response caused by
low cAMP levels, which may explain the observation that patients with T2DM are at an
increased risk of thrombosis [50]. Another study further established a relatively higher
degree of platelet inhibition by cilostazol in patients with diabetes compared with those
without this condition [51].
production of hepatocyte growth factor, which is known for its favorable role in attenuating
the malformation and apoptosis of endothelial cells [71].
Also, in a clinical setting, a three-month intervention using cilostazol improved en-
dothelial function, as measured by flow-mediated dilation, compared with aspirin in
patients with stroke (1.0% vs. 0.8%) [72]. Moreover, a six-month treatment with cilosta-
zol improved vasomotor reactivity, as evidenced by an increase in the coronary luminal
diameter following the administration of N-monomethyl-L-arginine [73].
Figure2.2.PRISMA
Figure PRISMAflow
flowdiagram
diagramfor
forstudies
studiesincluded
includedin
inthe
thecurrent
currentmeta-analysis.
meta-analysis.
All 33
All 33 studies
studies mentioned
mentionedabove abovewerewereincluded
includedin the meta-analysis
in the meta-analysis of cilostazol ther-
of cilostazol
apy for for
therapy thethe
prevention
prevention of of
composite
compositeMACE.
MACE.The Thedefinition
definitionofof MACE
MACE varied among among thethe
trials, which might be attributed to differences in patient characteristics
trials, which might be attributed to differences in patient characteristics (Supplementary (Supplementary
TableS2).
Table S2).Cilostazol
Cilostazoladministration
administrationeffectively
effectivelyreduced
reducedthetherisk
riskofofMACE
MACEcompared
comparedwithwith
boththe
both thecontrol
control(risk
(riskratio
ratio(RR):
(RR):0.62;
0.62;95%
95%CI:
CI:0.52–0.73)
0.52–0.73)(Figure
(Figure3A)3A) and
and aspirin
aspirin (RR:
(RR: 0.74;
0.74;
95% CI:
95% CI: 0.63–0.87) treatments
treatments (Figure
(Figure3B).
3B).Cilostazol
Cilostazolexhibited
exhibited a superior
a superior efficacy over
efficacy overas-
pirin forfor
aspirin secondary
secondary prevention,
prevention, whereas
whereasfurther
furtherevidence
evidenceis needed
is neededto draw a solid
to draw con-
a solid
clusion regarding
conclusion regardingits efficacy in primary
its efficacy in primaryprevention
prevention(Figure 3B).3B).
(Figure OurOurresults confirmed
results confirmedthe
favorable
the effects
favorable of of
effects cilostazol onon
cilostazol stroke,
stroke,which
whichareareeven
evenbetter
better than
than those of aspirin
aspirin [126]
[126]
(Figure
(Figure4B).
4B).The beneficial
The efficacy
beneficial of cilostazol
efficacy in reducing
of cilostazol the risk of
in reducing themyocardial infarction
risk of myocardial
and cardiovascular death was not demonstrated (Figure 4A,C). The distinction in responses
Int. J. Mol. Sci. 2024, 25, 2593 11 of 26
between ischemic stroke and myocardial infarction might lie in their differing associations
with underlying etiologies [127]. Ischemic stroke is primarily associated with the clogging
of blood vessels. In contrast, in myocardial infarction, multiple blood vessels may be
involved, and other risk factors often play a significant role before an attack occurs.
Cilostazol therapy yielded a reduced bleeding risk compared with aspirin (Figure 4E).
However, its usage was limited because of adverse events, leading to therapy discontin-
uation (Figure 4F), such as headache and palpitation (Supplementary Figure S3). This
suggests the need for improvements in this medication to address its side effects.
Table 2. Cont.
Table 2. Cont.
(A)
(B)
(C)
Figure
Figure 3. Forest plots of3.cilostazol
Forest plots of cilostazolmajor
in composite in composite
adversemajor adverse cardiovascular
cardiovascular events (MACEs)events (MAC
compared with thepared with (A)
following: thethe
following:
control, (A) the control,
(B) aspirin, (B) clopidogrel.
and (C) aspirin, andCI,
(C)confidence
clopidogrel. CI, confidence
interval;
RR, risk ratio. RR, risk ratio.
Int.Int. J. Mol.
J. Mol. Sci.
Sci. 2024,
2024, 25,25, x FOR PEER REVIEW
2593 1816ofof29
26
(A) (B)
(C) (D)
(E) (F)
Figure4.4.Forest
Figure Forestplots
plotsof
ofcilostazol
cilostazolin
incardiovascular
cardiovascular and
and safety
safety events: (A) myocardial
events: (A) myocardial infarction,
infarction,
(B)
(B)stroke,
stroke,(C)
(C)cardiovascular
cardiovasculardeath,
death,(D)
(D)all-cause
all-causedeath,
death, (E)
(E) bleeding, and (F) discontinuation
discontinuation of
of the
the
studydrug.
study drug.
4.5.
4.5.Peripheral
PeripheralArtery
ArteryDisease
Disease(PAD)
(PAD)
The
The clinical excellence ofcilostazol
clinical excellence of cilostazolisissufficient
sufficienttotorecommend
recommendit it asas
a first-line
a first-linedrug
drugin
PAD, because it increases NO in vascular endothelial cells in a dose-dependent
in PAD, because it increases NO in vascular endothelial cells in a dose-dependent manner, manner, to
dilate blood vessels [130]. A meta-analysis of 16 RCTs reported that intermittent
to dilate blood vessels [130]. A meta-analysis of 16 RCTs reported that intermittent clau- claudica-
tion was significantly
dication improved
was significantly by cilostazol
improved therapy
by cilostazol (100–300
therapy mg per
(100–300 mgday) compared
per day) comparedwith
the placebo [131]. An RCT including 698 patients with moderate-to-severe
with the placebo [131]. An RCT including 698 patients with moderate-to-severe claudica- claudication
symptoms
tion symptomsreported that that
reported a 6-month
a 6-monthcilostazol
cilostazol therapy
therapy improved
improvedthe themaximal walking
maximal walking
distance by 54%, compared with pentoxifylline therapy (30% increase)
distance by 54%, compared with pentoxifylline therapy (30% increase) [132]. Another [132]. Another study
including 394 patients with intermittent claudication showed that cilostazol
study including 394 patients with intermittent claudication showed that cilostazol treat- treatment for
6 ment
months improved the maximal walking distance compared with the
for 6 months improved the maximal walking distance compared with the placebo placebo [133]. A 2021
Cochrane Systematic
[133]. A 2021 CochraneReview and Meta-analysis
Systematic Review and including
Meta-analysis16 double-blind
including 16RCTs reported
double-blind
that
RCTsparticipants
reported that taking cilostazol
participants hadcilostazol
taking a higher had initial claudication
a higher distance, by
initial claudication 26.5 m
distance,
(95% CI: 18.9–34.1 m), compared with those taking the placebo [131].
by 26.5 m (95% CI: 18.9–34.1 m), compared with those taking the placebo [131]. Cilostazol Cilostazol exerted
beneficial effects regarding
exerted beneficial the primary
effects regarding the prevention of diabetic
primary prevention of foot ulcers
diabetic viaulcers
foot a lowering
via a
effect on MMP-9 levels [134].
lowering effect on MMP-9 levels [134].
Recently, a prospective study with 794 participants showed that the adjunctive ad-
ministration of cilostazol to clopidogrel-treated patients with T2DM with symptomatic
Int. J. Mol. Sci. 2024, 25, 2593 17 of 26
PAD lowered the risk of ischemic events and improved intermittent claudication symp-
toms, without increasing the bleeding risk [122]. Based on this evidence, the ACC/AHA
guidelines provided a class I recommendation for the use of cilostazol in the treatment of
intermittent claudication in 2016 and thereafter [135].
(A) (B)
(C) (D)
Figure
Figure 5. 5. Forest
Forest plots
plots of cilostazol’s
of cilostazol’s effect
effect on composite
on composite majormajor adverse
adverse cardiovascular
cardiovascular eventsevents
(MACEs)
(MACEs)
and and individual
individual components components
categorizedcategorized by regional
by regional background:
background: (A) MACE,
(A) MACE, (B) myocar-
(B) myocardial infarc-
dial infarction (MI), (C) stroke, and (D) cardiovascular death. CI, confidence interval; RR, risk ratio.
tion (MI), (C) stroke, and (D) cardiovascular death. CI, confidence interval; RR, risk ratio.
7. Conclusions
Cilostazol has multifaceted beneficial effects through various mechanisms, including
platelet inhibition, vasodilation via NO production, protective effects on vascular endo-
thelial cells, antiproliferative effects on VSMCs, and favorable changes in lipid profiles
(Figure 6). These beneficial effects of cilostazol are attributable to its pleiotropic properties,
Int. J. Mol. Sci. 2024, 25, 2593 19 of 26
7. Conclusions
Cilostazol has multifaceted beneficial effects through various mechanisms, includ-
Int. J. Mol. Sci. 2024, 25, x FOR PEERing
REVIEW inhibition, vasodilation via NO production, protective effects on 22
platelet of 29
vascular
endothelial cells, antiproliferative effects on VSMCs, and favorable changes in lipid profiles
(Figure 6). These beneficial effects of cilostazol are attributable to its pleiotropic properties,
including
includingits
itsanti-inflammatory
anti-inflammatory and antioxidative actions.AAneuroprotective
antioxidative actions. neuroprotective effect
effect waswas
also found in a few clinical studies of cilostazol.
also found in a few clinical studies cilostazol.
Basedon
Based onthe
therecent
recentresults
resultsofoflarge
largeclinical
clinicalstudies
studiessuggesting
suggestingthat
thataspirin
aspirinshould
shouldnotnotbe
be recommended in primary prevention settings, cilostazol treatment may
recommended in primary prevention settings, cilostazol treatment may be a viable option be a viable op-
tion
for for preventing
preventing the development
the development and progression
and progression of vascular
of vascular diseases.
diseases. Obviously,
Obviously, ci-
cilostazol
lostazol is effective in PAD, and recent evidence supports its effectiveness in CAD
is effective in PAD, and recent evidence supports its effectiveness in CAD as well as ischemic as well
as ischemic stroke. Notably, recent studies reported that cilostazol treatment was effica-
stroke. Notably, recent studies reported that cilostazol treatment was efficacious in reducing
cious in reducing atheromatous plaque progression in coronary arteries and decreasing
atheromatous plaque progression in coronary arteries and decreasing cardiovascular events
cardiovascular events compared with aspirin in patients with diabetes and subclinical
compared with aspirin in patients with diabetes and subclinical CAD. Thus, cilostazol
CAD. Thus, cilostazol might be helpful in the comprehensive vascular management of
might be helpful in the comprehensive vascular management of patients with a high risk
patients with a high risk of CV. However, it is noteworthy that these results were mainly
of CV. However, it is noteworthy that these results were mainly derived from East-Asian
derived from East-Asian populations. Additional trials including other ethnic groups are
populations. Additional trials including other ethnic groups are needed to generalize
needed to generalize the beneficial effects of this drug. Adverse events occurring after ci-
the beneficial effects of this drug. Adverse events occurring after cilostazol therapy, such
lostazol therapy, such as headache and heart rate increase, have not been completely re-
as headache and heart rate increase, have not been completely resolved; therefore, the
solved; therefore, the development of more-advanced PDE-3 inhibitors that do not exhibit
development of more-advanced PDE-3 inhibitors that do not exhibit these unwanted effects
these unwanted effects is warranted. Longer-term studies with a robust outcome are
is warranted. Longer-term studies with a robust outcome are needed to confirm the efficacy
needed to confirm the efficacy and safety of cilostazol therapy.
and safety of cilostazol therapy.
Supplementary Materials: The following supporting information can be downloaded at:
Supplementary Materials: The following supporting information can be downloaded at: https://www.
www.mdpi.com/xxx/s1.
mdpi.com/article/10.3390/ijms25052593/s1.
Author Contributions: Conceptualization, M.S. and S.L.; methodology, M.S. and S.L.; validation,
Author
M.S. andContributions: Conceptualization,
S.L.; formal analysis, M.S. and S.L.;M.S. and S.L.; M.S.
investigation, methodology, M.S. and M.S.
and S.L.; resources, S.L.; and
validation,
S.L.;
M.S.
dataand S.L.; formal
curation, analysis,
M.S. and M.S. and S.L.; investigation,
S.L.; writing—original M.S. and
draft preparation, M.S.S.L.;
andresources, M.S. and S.L.;
S.L.; writing—review
andcuration,
data editing, M.S.
M.S.andandS.L.;
S.L.;visualization, M.S. and
writing—original S.L.;
draft supervision,
preparation, S.L.;and
M.S. project
S.L.;administration,
writing—review S.L.
and
All authors
editing, M.S. have read visualization,
and S.L.; and agreed to M.S.
the published
and S.L.; version of theS.L.;
supervision, manuscript.
project administration, S.L. All
authors
Funding:have read
This and agreed
research to the
received published
no external version of the manuscript.
funding.
Funding: ThisReview
Institutional researchBoard
received no external
Statement: funding.
This is not applicable as this project did not involve humans
or animals.
Informed Consent Statement: Not applicable.
Data Availability Statement: The supporting data for the findings can be obtained from the corre-
sponding author upon a reasonable request.
Int. J. Mol. Sci. 2024, 25, 2593 20 of 26
Institutional Review Board Statement: This is not applicable as this project did not involve humans
or animals.
Informed Consent Statement: Not applicable.
Data Availability Statement: The supporting data for the findings can be obtained from the corre-
sponding author upon a reasonable request.
Conflicts of Interest: The authors declare no conflicts of interest.
References
1. Libby, P.; Buring, J.E.; Badimon, L.; Hansson, G.K.; Deanfield, J.; Bittencourt, M.S.; Tokgozoglu, L.; Lewis, E.F. Atherosclerosis.
Nat. Rev. Dis. Prim. 2019, 5, 56. [CrossRef]
2. Golledge, J. Update on the pathophysiology and medical treatment of peripheral artery disease. Nat. Rev. Cardiol. 2022, 19,
456–474. [CrossRef] [PubMed]
3. The Top 10 Causes of Death. Available online: https://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death
(accessed on 30 September 2023).
4. Wang, Y.; Li, Z.; Zhao, X.; Wang, D.; Li, H.; Xian, Y.; Liu, L.; Wang, Y. Stroke care quality in China: Substantial improvement, and a
huge challenge and opportunity. Int. J. Stroke 2017, 12, 229–235. [CrossRef]
5. Morley, R.L.; Sharma, A.; Horsch, A.D.; Hinchliffe, R.J. Peripheral artery disease. BMJ 2018, 360, j5842. [CrossRef] [PubMed]
6. Mollon, L.; Bhattacharjee, S. Health related quality of life among myocardial infarction survivors in the United States: A propensity
score matched analysis. Health Qual. Life Outcomes 2017, 15, 235. [CrossRef]
7. Sprigg, N.; Selby, J.; Fox, L.; Berge, E.; Whynes, D.; Bath, P.M.; Efficacy of Nitric Oxide in Stroke Investigators. Very low quality of
life after acute stroke: Data from the Efficacy of Nitric Oxide in Stroke trial. Stroke 2013, 44, 3458–3462. [CrossRef]
8. Jouven, X.; Lemaitre, R.N.; Rea, T.D.; Sotoodehnia, N.; Empana, J.P.; Siscovick, D.S. Diabetes, glucose level, and risk of sudden
cardiac death. Eur. Heart J. 2005, 26, 2142–2147. [CrossRef]
9. Emerging Risk Factors, C.; Sarwar, N.; Gao, P.; Seshasai, S.R.; Gobin, R.; Kaptoge, S.; Di Angelantonio, E.; Ingelsson, E.; Lawlor,
D.A.; Selvin, E.; et al. Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: A collaborative
meta-analysis of 102 prospective studies. Lancet 2010, 375, 2215–2222. [CrossRef]
10. Beckman, J.A.; Creager, M.A. Vascular Complications of Diabetes. Circ. Res. 2016, 118, 1771–1785. [CrossRef] [PubMed]
11. Low Wang, C.C.; Hess, C.N.; Hiatt, W.R.; Goldfine, A.B. Clinical Update: Cardiovascular Disease in Diabetes Mellitus: Atheroscle-
rotic Cardiovascular Disease and Heart Failure in Type 2 Diabetes Mellitus—Mechanisms, Management, and Clinical Considera-
tions. Circulation 2016, 133, 2459–2502. [CrossRef] [PubMed]
12. Patel, K.V.; Pandey, A.; de Lemos, J.A. Conceptual Framework for Addressing Residual Atherosclerotic Cardiovascular Disease
Risk in the Era of Precision Medicine. Circulation 2018, 137, 2551–2553. [CrossRef]
13. ElSayed, N.A.; Aleppo, G.; Aroda, V.R.; Bannuru, R.R.; Brown, F.M.; Bruemmer, D.; Collins, B.S.; Das, S.R.; Hilliard, M.E.;
Isaacs, D.; et al. 10. Cardiovascular Disease and Risk Management: Standards of Care in Diabetes—2023. Diabetes Care 2023, 46
(Suppl. S1), S158–S190. [CrossRef]
14. Marx, N.; Federici, M.; Schutt, K.; Muller-Wieland, D.; Ajjan, R.A.; Antunes, M.J.; Christodorescu, R.M.; Crawford, C.; Di
Angelantonio, E.; Eliasson, B.; et al. 2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes.
Eur. Heart J. 2023, 44, 4043–4140. [CrossRef] [PubMed]
15. Kambayashi, J.; Liu, Y.; Sun, B.; Shakur, Y.; Yoshitake, M.; Czerwiec, F. Cilostazol as a unique antithrombotic agent. Curr. Pharm.
Des. 2003, 9, 2289–2302. [CrossRef] [PubMed]
16. Degerman, E.; Belfrage, P.; Manganiello, V.C. Structure, localization, and regulation of cGMP-inhibited phosphodiesterase (PDE3).
J. Biol. Chem. 1997, 272, 6823–6826. [CrossRef] [PubMed]
17. Huh, J.H.; Seok, H.; Lee, B.W.; Kang, E.S.; Lee, H.C.; Cha, B.S. Effect of cilostazol on carotid intima-media thickness in type 2
diabetic patients without cardiovascular event. Endocrine 2014, 47, 138–145. [CrossRef] [PubMed]
18. Katakami, N.; Kim, Y.S.; Kawamori, R.; Yamasaki, Y. The phosphodiesterase inhibitor cilostazol induces regression of carotid
atherosclerosis in subjects with type 2 diabetes mellitus: Principal results of the Diabetic Atherosclerosis Prevention by Cilostazol
(DAPC) study: A randomized trial. Circulation 2010, 121, 2584–2591. [CrossRef] [PubMed]
19. Kwon, S.U.; Cho, Y.J.; Koo, J.S.; Bae, H.J.; Lee, Y.S.; Hong, K.S.; Lee, J.H.; Kim, J.S. Cilostazol prevents the progression of the
symptomatic intracranial arterial stenosis: The multicenter double-blind placebo-controlled trial of cilostazol in symptomatic
intracranial arterial stenosis. Stroke 2005, 36, 782–786. [CrossRef] [PubMed]
20. Shinohara, Y.; Katayama, Y.; Uchiyama, S.; Yamaguchi, T.; Handa, S.; Matsuoka, K.; Ohashi, Y.; Tanahashi, N.; Yamamoto,
H.; Genka, C.; et al. Cilostazol for prevention of secondary stroke (CSPS 2): An aspirin-controlled, double-blind, randomised
non-inferiority trial. Lancet Neurol. 2010, 9, 959–968. [CrossRef]
21. Biondi-Zoccai, G.G.; Lotrionte, M.; Anselmino, M.; Moretti, C.; Agostoni, P.; Testa, L.; Abbate, A.; Cosgrave, J.; Laudito, A.;
Trevi, G.P.; et al. Systematic review and meta-analysis of randomized clinical trials appraising the impact of cilostazol after
percutaneous coronary intervention. Am. Heart J. 2008, 155, 1081–1089. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 2593 21 of 26
22. Angiolillo, D.J.; Capranzano, P.; Goto, S.; Aslam, M.; Desai, B.; Charlton, R.K.; Suzuki, Y.; Box, L.C.; Shoemaker, S.B.; Zenni,
M.M.; et al. A randomized study assessing the impact of cilostazol on platelet function profiles in patients with diabetes mellitus
and coronary artery disease on dual antiplatelet therapy: Results of the OPTIMUS-2 study. Eur. Heart J. 2008, 29, 2202–2211.
[CrossRef]
23. Rogers, K.C.; Oliphant, C.S.; Finks, S.W. Clinical efficacy and safety of cilostazol: A critical review of the literature. Drugs 2015, 75,
377–395. [CrossRef]
24. Berger, J.S. Aspirin for Primary Prevention-Time to Rethink Our Approach. JAMA Netw. Open 2022, 5, e2210144. [CrossRef]
25. Ogawa, H.; Nakayama, M.; Morimoto, T.; Uemura, S.; Kanauchi, M.; Doi, N.; Jinnouchi, H.; Sugiyama, S.; Saito, Y.; Japanese
Primary Prevention of Atherosclerosis With Aspirin for Diabetes (JPAD) Trial Investigators. Low-dose aspirin for primary
prevention of atherosclerotic events in patients with type 2 diabetes: A randomized controlled trial. JAMA 2008, 300, 2134–2141.
[CrossRef]
26. Belch, J.; MacCuish, A.; Campbell, I.; Cobbe, S.; Taylor, R.; Prescott, R.; Lee, R.; Bancroft, J.; MacEwan, S.; Shepherd, J.; et al. The
prevention of progression of arterial disease and diabetes (POPADAD) trial: Factorial randomised placebo controlled trial of
aspirin and antioxidants in patients with diabetes and asymptomatic peripheral arterial disease. BMJ 2008, 337, a1840. [CrossRef]
[PubMed]
27. Calvin, A.D.; Aggarwal, N.R.; Murad, M.H.; Shi, Q.; Elamin, M.B.; Geske, J.B.; Fernandez-Balsells, M.M.; Albuquerque, F.N.;
Lampropulos, J.F.; Erwin, P.J.; et al. Aspirin for the primary prevention of cardiovascular events: A systematic review and
meta-analysis comparing patients with and without diabetes. Diabetes Care 2009, 32, 2300–2306. [CrossRef] [PubMed]
28. Ikeda, Y.; Shimada, K.; Teramoto, T.; Uchiyama, S.; Yamazaki, T.; Oikawa, S.; Sugawara, M.; Ando, K.; Murata, M.; Yokoyama, K.;
et al. Low-dose aspirin for primary prevention of cardiovascular events in Japanese patients 60 years or older with atherosclerotic
risk factors: A randomized clinical trial. JAMA 2014, 312, 2510–2520. [CrossRef] [PubMed]
29. de Gaetano, G.; Collaborative Group of the Primary Prevention Project. Low-dose aspirin and vitamin E in people at cardiovascu-
lar risk: A randomised trial in general practice. Collaborative Group of the Primary Prevention Project. Lancet 2001, 357, 89–95.
[PubMed]
30. Ridker, P.M.; Cook, N.R.; Lee, I.M.; Gordon, D.; Gaziano, J.M.; Manson, J.E.; Hennekens, C.H.; Buring, J.E. A randomized trial of
low-dose aspirin in the primary prevention of cardiovascular disease in women. N. Engl. J. Med. 2005, 352, 1293–1304. [CrossRef]
[PubMed]
31. Group, A.S.C.; Bowman, L.; Mafham, M.; Wallendszus, K.; Stevens, W.; Buck, G.; Barton, J.; Murphy, K.; Aung, T.; Haynes, R.;
et al. Effects of Aspirin for Primary Prevention in Persons with Diabetes Mellitus. N. Engl. J. Med. 2018, 379, 1529–1539.
32. Gaziano, J.M.; Brotons, C.; Coppolecchia, R.; Cricelli, C.; Darius, H.; Gorelick, P.B.; Howard, G.; Pearson, T.A.; Rothwell, P.M.;
Ruilope, L.M.; et al. Use of aspirin to reduce risk of initial vascular events in patients at moderate risk of cardiovascular disease
(ARRIVE): A randomised, double-blind, placebo-controlled trial. Lancet 2018, 392, 1036–1046. [CrossRef]
33. McNeil, J.J.; Nelson, M.R.; Woods, R.L.; Lockery, J.E.; Wolfe, R.; Reid, C.M.; Kirpach, B.; Shah, R.C.; Ives, D.G.; Storey, E.; et al.
Effect of Aspirin on All-Cause Mortality in the Healthy Elderly. N. Engl. J. Med. 2018, 379, 1519–1528. [CrossRef]
34. Yusuf, S.; Joseph, P.; Dans, A.; Gao, P.; Teo, K.; Xavier, D.; Lopez-Jaramillo, P.; Yusoff, K.; Santoso, A.; Gamra, H.; et al. Polypill
with or without Aspirin in Persons without Cardiovascular Disease. N. Engl. J. Med. 2021, 384, 216–228. [CrossRef]
35. Massimi, I.; Lotti, L.V.; Temperilli, F.; Mancone, M.; Sardella, G.; Calcagno, S.; Turriziani, O.; Frati, L.; Pulcinelli, F.M. Enhanced
platelet MRP4 expression and correlation with platelet function in patients under chronic aspirin treatment. Thromb. Haemost.
2016, 116, 1100–1110. [CrossRef] [PubMed]
36. Angiolillo, D.J. Variability in responsiveness to oral antiplatelet therapy. Am. J. Cardiol. 2009, 103 (Suppl. S3), 27A–34A. [CrossRef]
[PubMed]
37. Mora, S.; Shufelt, C.L.; Manson, J.E. Whom to Treat for Primary Prevention of Atherosclerotic Cardiovascular Disease: The
Aspirin Dilemma. JAMA Intern. Med. 2022, 182, 587–589. [CrossRef]
38. American Diabetes, A. 8. Cardiovascular Disease and Risk Management. Diabetes Care 2016, 39 (Suppl. S1), S60–S71. [CrossRef]
39. Pignone, M.; Alberts, M.J.; Colwell, J.A.; Cushman, M.; Inzucchi, S.E.; Mukherjee, D.; Rosenson, R.S.; Williams, C.D.; Wilson, P.W.;
Kirkman, M.S.; et al. Aspirin for primary prevention of cardiovascular events in people with diabetes: A position statement of
the American Diabetes Association, a scientific statement of the American Heart Association, and an expert consensus document
of the American College of Cardiology Foundation. Diabetes Care 2010, 33, 1395–1402. [PubMed]
40. Xue, Y.; Wang, Z.; Wu, H.; Li, X.; Chen, J.; Lv, Q. Cilostazol increases adenosine plasma concentration in patients with acute
coronary syndrome. J. Clin. Pharm. Ther. 2021, 46, 328–332. [CrossRef]
41. Yun, S.H.; Sim, E.H.; Goh, R.Y.; Park, J.I.; Han, J.Y. Platelet Activation: The Mechanisms and Potential Biomarkers. Biomed. Res.
Int. 2016, 2016, 9060143. [CrossRef]
42. Paul, S.; Feoktistov, I.; Hollister, A.S.; Robertson, D.; Biaggioni, I. Adenosine inhibits the rise in intracellular calcium and platelet
aggregation produced by thrombin: Evidence that both effects are coupled to adenylate cyclase. Mol. Pharmacol. 1990, 37, 870–875.
43. Yan, R.; Li, S.; Dai, K. The critical roles of cyclic AMP/cyclic AMP-dependent protein kinase in platelet physiology. Front. Biol.
China 2009, 4, 7–14. [CrossRef]
44. Guarino, M.L.; Massimi, I.; Alemanno, L.; Conti, L.; Angiolillo, D.J.; Pulcinelli, F.M. MRP4 over-expression has a role on both
reducing nitric oxide-dependent antiplatelet effect and enhancing ADP induced platelet activation. J. Thromb. Thrombolysis 2021,
51, 625–632. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 2593 22 of 26
45. Alemanno, L.; Massimi, I.; Klaus, V.; Guarino, M.L.; Maltese, T.; Frati, L.; Angiolillo, D.J.; Pulcinelli, F.M. Impact of Multidrug
Resistance Protein-4 Inhibitors on Modulating Platelet Function and High on-Aspirin Treatment Platelet Reactivity. Thromb.
Haemost. 2018, 118, 490–501. [CrossRef]
46. Cheepala, S.B.; Pitre, A.; Fukuda, Y.; Takenaka, K.; Zhang, Y.; Wang, Y.; Frase, S.; Pestina, T.; Gartner, T.K.; Jackson, C.; et al. The
ABCC4 membrane transporter modulates platelet aggregation. Blood 2015, 126, 2307–2319. [CrossRef] [PubMed]
47. Borgognone, A.; Pulcinelli, F.M. Reduction of cAMP and cGMP inhibitory effects in human platelets by MRP4-mediated transport.
Thromb. Haemost. 2012, 108, 955–962. [CrossRef] [PubMed]
48. Ohnuki, Y.; Ohnuki, Y.; Kohara, S.; Shimizu, M.; Takizawa, S. Dual Therapy with Aspirin and Cilostazol May Improve Platelet
Aggregation in Noncardioembolic Stroke Patients: A Pilot Study. Intern. Med. 2017, 56, 1307–1313. [CrossRef]
49. Jeong, Y.H.; Hwang, J.Y.; Kim, I.S.; Park, Y.; Hwang, S.J.; Lee, S.W.; Kwak, C.H.; Park, S.W. Adding cilostazol to dual antiplatelet
therapy achieves greater platelet inhibition than high maintenance dose clopidogrel in patients with acute myocardial infarction:
Results of the adjunctive cilostazol versus high maintenance dose clopidogrel in patients with AMI (ACCEL-AMI) study. Circ.
Cardiovasc. Interv. 2010, 3, 17–26.
50. Angiolillo, D.J. Antiplatelet therapy in type 2 diabetes mellitus. Curr. Opin. Endocrinol. Diabetes Obes. 2007, 14, 124–131. [CrossRef]
[PubMed]
51. Capranzano, P.; Ferreiro, J.L.; Ueno, M.; Capodanno, D.; Dharmashankar, K.; Darlington, A.; Desai, B.; Tello-Montoliu, A.; Rollini,
F.; Angiolillo, D.J. Pharmacodynamic effects of adjunctive cilostazol therapy in patients with coronary artery disease on dual
antiplatelet therapy: Impact of high on-treatment platelet reactivity and diabetes mellitus status. Catheter. Cardiovasc. Interv. 2013,
81, 42–49. [CrossRef]
52. Manickavasagam, S.; Ye, Y.; Lin, Y.; Perez-Polo, R.J.; Huang, M.H.; Lui, C.Y.; Hughes, M.G.; McAdoo, D.J.; Uretsky, B.F.; Birnbaum,
Y. The cardioprotective effect of a statin and cilostazol combination: Relationship to Akt and endothelial nitric oxide synthase
activation. Cardiovasc. Drugs Ther. 2007, 21, 321–330. [CrossRef] [PubMed]
53. Tanaka, T.; Ishikawa, T.; Hagiwara, M.; Onoda, K.; Itoh, H.; Hidaka, H. Effects of cilostazol, a selective cAMP phosphodiesterase
inhibitor on the contraction of vascular smooth muscle. Pharmacology 1988, 36, 313–320. [CrossRef] [PubMed]
54. Wu, S.N.; Liu, S.I.; Huang, M.H. Cilostazol, an inhibitor of type 3 phosphodiesterase, stimulates large-conductance, calcium-
activated potassium channels in pituitary GH3 cells and pheochromocytoma PC12 cells. Endocrinology 2004, 145, 1175–1184.
[CrossRef] [PubMed]
55. Dindyal, S.; Kyriakides, C. A review of cilostazol, a phosphodiesterase inhibitor, and its role in preventing both coronary and
peripheral arterial restenosis following endovascular therapy. Recent. Pat. Cardiovasc. Drug Discov. 2009, 4, 6–14. [CrossRef]
[PubMed]
56. Nakamura, T.; Houchi, H.; Minami, A.; Sakamoto, S.; Tsuchiya, K.; Niwa, Y.; Minakuchi, K.; Nakaya, Y. Endothelium-dependent
relaxation by cilostazol, a phosphodiesteras III inhibitor, on rat thoracic aorta. Life Sci. 2001, 69, 1709–1715. [CrossRef]
57. Li, H.; Hong, D.H.; Son, Y.K.; Na, S.H.; Jung, W.K.; Bae, Y.M.; Seo, E.Y.; Kim, S.J.; Choi, I.W.; Park, W.S. Cilostazol induces
vasodilation through the activation of Ca(2+) -activated K(+) channels in aortic smooth muscle. Vasc. Pharmacol. 2015, 70, 15–22.
[CrossRef]
58. Liu, Y.; Shakur, Y.; Yoshitake, M.; Kambayashi Ji, J. Cilostazol (pletal): A dual inhibitor of cyclic nucleotide phosphodiesterase
type 3 and adenosine uptake. Cardiovasc. Drug Rev. 2001, 19, 369–386. [CrossRef]
59. Liu, Y.; Fong, M.; Cone, J.; Wang, S.; Yoshitake, M.; Kambayashi, J. Inhibition of adenosine uptake and augmentation of ischemia-
induced increase of interstitial adenosine by cilostazol, an agent to treat intermittent claudication. J. Cardiovasc. Pharmacol. 2000,
36, 351–360. [CrossRef]
60. Shryock, J.C.; Belardinelli, L. Adenosine and adenosine receptors in the cardiovascular system: Biochemistry, physiology, and
pharmacology. Am. J. Cardiol. 1997, 79, 2–10. [CrossRef]
61. Chancharoenthana, W.; Leelahavanichkul, A.; Taratummarat, S.; Wongphom, J.; Tiranathanagul, K.; Eiam-Ong, S. Cilostazol
attenuates intimal hyperplasia in a mouse model of chronic kidney disease. PLoS ONE 2017, 12, e0187872. [CrossRef]
62. Takahashi, S.; Oida, K.; Fujiwara, R.; Maeda, H.; Hayashi, S.; Takai, H.; Tamai, T.; Nakai, T.; Miyabo, S. Effect of cilostazol, a cyclic
AMP phosphodiesterase inhibitor, on the proliferation of rat aortic smooth muscle cells in culture. J. Cardiovasc. Pharmacol. 1992,
20, 900–906. [CrossRef]
63. Kim, M.J.; Park, K.G.; Lee, K.M.; Kim, H.S.; Kim, S.Y.; Kim, C.S.; Lee, S.L.; Chang, Y.C.; Park, J.Y.; Lee, K.U.; et al. Cilostazol
inhibits vascular smooth muscle cell growth by downregulation of the transcription factor E2F. Hypertension 2005, 45, 552–556.
[CrossRef]
64. Takigawa, T.; Tsurushima, H.; Suzuki, K.; Tsuruta, W.; Nakamura, K.; Matsumura, A. Cilostazol suppression of arterial intimal
hyperplasia is associated with decreased expression of sialyl Lewis X homing receptors on mononuclear cells and E-selectin in
endothelial cells. J. Vasc. Surg. 2012, 55, 506–516. [CrossRef] [PubMed]
65. Omi, H.; Okayama, N.; Shimizu, M.; Fukutomi, T.; Nakamura, A.; Imaeda, K.; Okouchi, M.; Itoh, M. Cilostazol inhibits high
glucose-mediated endothelial-neutrophil adhesion by decreasing adhesion molecule expression via NO production. Microvasc.
Res. 2004, 68, 119–125. [CrossRef]
66. Deglise, S.; Bechelli, C.; Allagnat, F. Vascular smooth muscle cells in intimal hyperplasia, an update. Front. Physiol. 2022, 13,
1081881. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 2593 23 of 26
67. Chen, W.J.; Chen, Y.H.; Lin, K.H.; Ting, C.H.; Yeh, Y.H. Cilostazol promotes vascular smooth muscles cell differentiation through
the cAMP response element-binding protein-dependent pathway. Arter. Thromb. Vasc. Biol. 2011, 31, 2106–2113. [CrossRef]
[PubMed]
68. Hashimoto, A.; Miyakoda, G.; Hirose, Y.; Mori, T. Activation of endothelial nitric oxide synthase by cilostazol via a cAMP/protein
kinase A- and phosphatidylinositol 3-kinase/Akt-dependent mechanism. Atherosclerosis 2006, 189, 350–357. [CrossRef]
69. Moreira, H.S.; Lima-Leal, G.A.; Santos-Rocha, J.; Gomes-Pereira, L.; Duarte, G.P.; Xavier, F.E. Phosphodiesterase-3 inhibitor
cilostazol reverses endothelial dysfunction with ageing in rat mesenteric resistance arteries. Eur. J. Pharmacol. 2018, 822, 59–68.
[CrossRef] [PubMed]
70. Suzuki, K.; Uchida, K.; Nakanishi, N.; Hattori, Y. Cilostazol activates AMP-activated protein kinase and restores endothelial
function in diabetes. Am. J. Hypertens. 2008, 21, 451–457. [CrossRef] [PubMed]
71. Morishita, R.; Higaki, J.; Hayashi, S.I.; Yo, Y.; Aoki, M.; Nakamura, S.; Moriguchi, A.; Matsushita, H.; Matsumoto, K.; Nakamura,
T.; et al. Role of hepatocyte growth factor in endothelial regulation: Prevention of high D-glucose-induced endothelial cell death
by prostaglandins and phosphodiesterase type 3 inhibitor. Diabetologia 1997, 40, 1053–1061. [CrossRef]
72. Lee, S.J.; Lee, J.S.; Choi, M.H.; Lee, S.E.; Shin, D.H.; Hong, J.M. Cilostazol improves endothelial function in acute cerebral ischemia
patients: A double-blind placebo controlled trial with flow-mediated dilation technique. BMC Neurol. 2017, 17, 169. [CrossRef]
73. Watanabe, K.; Ikeda, S.; Komatsu, J.; Inaba, S.; Suzuki, J.; Sueda, S.; Funada, J.; Kitakaze, M.; Sekiya, M. Effect of cilostazol on
vasomotor reactivity in patients with vasospastic angina pectoris. Am. J. Cardiol. 2003, 92, 21–25. [CrossRef]
74. Zaki, O.S.; Nassar, N.N.; Abdallah, D.M.; Safar, M.M.; Mohammed, R.A. Cilostazol Alleviates NLRP3 Inflammasome-
Induced Allodynia/Hyperalgesia in Murine Cerebral Cortex Following Transient Ischemia: Focus on TRPA1/Glutamate and
Akt/Dopamine/BDNF/Nrf2 Trajectories. Mol. Neurobiol. 2022, 59, 7194–7211. [CrossRef]
75. Hoseini, Z.; Sepahvand, F.; Rashidi, B.; Sahebkar, A.; Masoudifar, A.; Mirzaei, H. NLRP3 inflammasome: Its regulation and
involvement in atherosclerosis. J. Cell Physiol. 2018, 233, 2116–2132. [CrossRef] [PubMed]
76. Grebe, A.; Hoss, F.; Latz, E. NLRP3 Inflammasome and the IL-1 Pathway in Atherosclerosis. Circ. Res. 2018, 122, 1722–1740.
[CrossRef] [PubMed]
77. de Oliveira Lopes, R.; Lima, G.F.; Mendes, A.B.A.; Autran, L.J.; de Assis Pereira, N.C.; Brazao, S.C.; Alexandre-Santos, B.;
Frantz, E.D.C.; Scaramello, C.B.V.; Brito, F.C.F.; et al. Cilostazol attenuates cardiac oxidative stress and inflammation in
hypercholesterolemic rats. Naunyn Schmiedebergs Arch. Pharmacol. 2022, 395, 789–801. [CrossRef] [PubMed]
78. Li, J.; Xiang, X.; Gong, X.; Shi, Y.; Yang, J.; Xu, Z. Cilostazol protects mice against myocardium ischemic/reperfusion injury by
activating a PPARgamma/JAK2/STAT3 pathway. Biomed. Pharmacother. 2017, 94, 995–1001. [CrossRef] [PubMed]
79. Agrawal, N.K.; Maiti, R.; Dash, D.; Pandey, B.L. Cilostazol reduces inflammatory burden and oxidative stress in hypertensive
type 2 diabetes mellitus patients. Pharmacol. Res. 2007, 56, 118–123. [CrossRef] [PubMed]
80. Lee, D.H.; Chun, E.J.; Oh, T.J.; Kim, K.M.; Moon, J.H.; Choi, S.H.; Park, K.S.; Jang, H.C.; Lim, S. Effect of cilostazol, a
phosphodiesterase-3 inhibitor, on coronary artery stenosis and plaque characteristics in patients with type 2 diabetes: ES-
CAPE study. Diabetes Obes. Metab. 2019, 21, 1409–1418. [CrossRef] [PubMed]
81. Ahn, C.M.; Hong, S.J.; Park, J.H.; Kim, J.S.; Lim, D.S. Cilostazol reduces the progression of carotid intima-media thickness without
increasing the risk of bleeding in patients with acute coronary syndrome during a 2-year follow-up. Heart Vessel. 2011, 26, 502–510.
[CrossRef]
82. Gendy, A.M.; Amin, M.M.; Al-Mokaddem, A.K.; Abd Ellah, M.F. Cilostazol mitigates mesenteric ischemia/reperfusion-induced
lung lesion: Contribution of PPAR-gamma, NF-kappaB, and STAT3 crosstalk. Life Sci. 2021, 266, 118882. [CrossRef]
83. Fujii, T.; Obara, H.; Matsubara, K.; Fujimura, N.; Yagi, H.; Hibi, T.; Abe, Y.; Kitago, M.; Shinoda, M.; Itano, O.; et al. Oral
administration of cilostazol improves survival rate after rat liver ischemia/reperfusion injury. J. Surg. Res. 2017, 213, 207–214.
[CrossRef]
84. Bai, Y.; Muqier; Murakami, H.; Iwasa, M.; Sumi, S.; Yamada, Y.; Ushikoshi, H.; Aoyama, T.; Nishigaki, K.; Takemura, G.; et al.
Cilostazol protects the heart against ischaemia reperfusion injury in a rabbit model of myocardial infarction: Focus on adenosine,
nitric oxide and mitochondrial ATP-sensitive potassium channels. Clin. Exp. Pharmacol. Physiol. 2011, 38, 658–665. [CrossRef]
85. Tani, T.; Uehara, K.; Sudo, T.; Marukawa, K.; Yasuda, Y.; Kimura, Y. Cilostazol, a selective type III phosphodiesterase inhibitor,
decreases triglyceride and increases HDL cholesterol levels by increasing lipoprotein lipase activity in rats. Atherosclerosis 2000,
152, 299–305. [CrossRef] [PubMed]
86. Hirose, T.; Kurebayashi, S.; Kasayama, S. Antiplatelet agent cilostazol potentiates adipocyte differentiation of 3T3-L1 cells.
Atherosclerosis 2001, 158, 19–22. [CrossRef] [PubMed]
87. Kim, H.J.; Moon, J.H.; Kim, H.M.; Yun, M.R.; Jeon, B.H.; Lee, B.; Kang, E.S.; Lee, H.C.; Cha, B.S. The hypolipidemic effect
of cilostazol can be mediated by regulation of hepatic low-density lipoprotein receptor-related protein 1 (LRP1) expression.
Metabolism 2014, 63, 112–119. [CrossRef] [PubMed]
88. Oldoni, F.; van Capelleveen, J.C.; Dalila, N.; Wolters, J.C.; Heeren, J.; Sinke, R.J.; Hui, D.Y.; Dallinga-Thie, G.M.; Frikke-Schmidt,
R.; Hovingh, K.G.; et al. Naturally Occurring Variants in LRP1 (Low-Density Lipoprotein Receptor-Related Protein 1) Affect HDL
(High-Density Lipoprotein) Metabolism Through ABCA1 (ATP-Binding Cassette A1) and SR-B1 (Scavenger Receptor Class B
Type 1) in Humans. Arter. Thromb. Vasc. Biol. 2018, 38, 1440–1453. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 2593 24 of 26
89. Ito, H.; Uehara, K.; Matsumoto, Y.; Hashimoto, A.; Nagano, C.; Niimi, M.; Miyakoda, G.; Nagano, K. Cilostazol inhibits
accumulation of triglyceride in aorta and platelet aggregation in cholesterol-fed rabbits. PLoS ONE 2012, 7, e39374. [CrossRef]
[PubMed]
90. da Motta, N.A.; de Brito, F.C. Cilostazol exerts antiplatelet and anti-inflammatory effects through AMPK activation and NF-kB
inhibition on hypercholesterolemic rats. Fundam. Clin. Pharmacol. 2016, 30, 327–337. [CrossRef] [PubMed]
91. Seo, D.H.; Shin, E.; Lee, Y.H.; Park, S.E.; Nam, K.T.; Kim, J.W.; Cha, B.S. Effects of a Phosphodiesterase inhibitor on the Browning
of Adipose Tissue in Mice. Biomedicines 2022, 10, 1852. [CrossRef] [PubMed]
92. Nedergaard, J.; Golozoubova, V.; Matthias, A.; Asadi, A.; Jacobsson, A.; Cannon, B. UCP1: The only protein able to mediate
adaptive non-shivering thermogenesis and metabolic inefficiency. Biochim. Biophys. Acta 2001, 1504, 82–106. [CrossRef]
93. Thompson, P.D.; Zimet, R.; Forbes, W.P.; Zhang, P. Meta-analysis of results from eight randomized, placebo-controlled trials on
the effect of cilostazol on patients with intermittent claudication. Am. J. Cardiol. 2002, 90, 1314–1319. [CrossRef]
94. Rizzo, M.; Corrado, E.; Patti, A.M.; Rini, G.B.; Mikhailidis, D.P. Cilostazol and atherogenic dyslipidemia: A clinically relevant
effect? Expert. Opin. Pharmacother. 2011, 12, 647–655. [CrossRef]
95. Gotoh, F.; Tohgi, H.; Hirai, S.; Terashi, A.; Fukuuchi, Y.; Otomo, E.; Shinohara, Y.; Itoh, E.; Matsuda, T.; Sawada, T.; et al.
Cilostazol stroke prevention study: A placebo-controlled double-blind trial for secondary prevention of cerebral infarction. J.
Stroke Cerebrovasc. Dis. 2000, 9, 147–157. [CrossRef]
96. Douglas, J.S., Jr.; Holmes, D.R., Jr.; Kereiakes, D.J.; Grines, C.L.; Block, E.; Ghazzal, Z.M.; Morris, D.C.; Liberman, H.; Parker, K.;
Jurkovitz, C.; et al. Coronary stent restenosis in patients treated with cilostazol. Circulation 2005, 112, 2826–2832. [CrossRef]
[PubMed]
97. Chen, Y.D.; Lu, Y.L.; Jin, Z.N.; Yuan, F.; Lu, S.Z. A prospective randomized antiplatelet trial of cilostazol versus clopidogrel in
patients with bare metal stent. Chin. Med. J. 2006, 119, 360–366. [CrossRef] [PubMed]
98. Lee, S.W.; Park, S.W.; Kim, Y.H.; Yun, S.C.; Park, D.W.; Lee, C.W.; Hong, M.K.; Kim, H.S.; Ko, J.K.; Park, J.H.; et al. Comparison of
triple versus dual antiplatelet therapy after drug-eluting stent implantation (from the DECLARE-Long trial). Am. J. Cardiol. 2007,
100, 1103–1108. [CrossRef] [PubMed]
99. Hiatt, W.R.; Money, S.R.; Brass, E.P. Long-term safety of cilostazol in patients with peripheral artery disease: The CASTLE study
(Cilostazol: A Study in Long-term Effects). J. Vasc. Surg. 2008, 47, 330–336. [CrossRef] [PubMed]
100. Huang, Y.; Cheng, Y.; Wu, J.; Li, Y.; Xu, E.; Hong, Z.; Li, Z.; Zhang, W.; Ding, M.; Gao, X.; et al. Cilostazol as an alternative to
aspirin after ischaemic stroke: A randomised, double-blind, pilot study. Lancet Neurol. 2008, 7, 494–499. [CrossRef] [PubMed]
101. Lee, S.W.; Park, S.W.; Kim, Y.H.; Yun, S.C.; Park, D.W.; Lee, C.W.; Hong, M.K.; Kim, H.S.; Ko, J.K.; Park, J.H.; et al. Drug-eluting
stenting followed by cilostazol treatment reduces late restenosis in patients with diabetes mellitus the DECLARE-DIABETES Trial
(A Randomized Comparison of Triple Antiplatelet Therapy with Dual Antiplatelet Therapy after Drug-Eluting Stent Implantation
in Diabetic Patients). J. Am. Coll. Cardiol. 2008, 51, 1181–1187. [PubMed]
102. Guo, J.J.; Xu, E.; Lin, Q.Y.; Zeng, G.L.; Xie, H.F. Effect of cilostazol on cerebral arteries in secondary prevention of ischemic stroke.
Neurosci. Bull. 2009, 25, 383–390. [CrossRef] [PubMed]
103. Han, Y.; Li, Y.; Wang, S.; Jing, Q.; Wang, Z.; Wang, D.; Shu, Q.; Tang, X. Cilostazol in addition to aspirin and clopidogrel improves
long-term outcomes after percutaneous coronary intervention in patients with acute coronary syndromes: A randomized,
controlled study. Am. Heart J. 2009, 157, 733–739. [CrossRef]
104. Soga, Y.; Yokoi, H.; Kawasaki, T.; Nakashima, H.; Tsurugida, M.; Hikichi, Y.; Nobuyoshi, M. Efficacy of cilostazol after endovascular
therapy for femoropopliteal artery disease in patients with intermittent claudication. J. Am. Coll. Cardiol. 2009, 53, 48–53.
[CrossRef] [PubMed]
105. Kwon, S.U.; Hong, K.S.; Kang, D.W.; Park, J.M.; Lee, J.H.; Cho, Y.J.; Yu, K.H.; Koo, J.S.; Wong, K.S.; Lee, S.H.; et al. Efficacy and
safety of combination antiplatelet therapies in patients with symptomatic intracranial atherosclerotic stenosis. Stroke 2011, 42,
2883–2890. [CrossRef]
106. Lee, S.W.; Park, S.W.; Kim, Y.H.; Yun, S.C.; Park, D.W.; Lee, C.W.; Kang, S.J.; Park, S.J.; Lee, J.H.; Choi, S.W.; et al. A randomized,
double-blind, multicenter comparison study of triple antiplatelet therapy with dual antiplatelet therapy to reduce restenosis after
drug-eluting stent implantation in long coronary lesions: Results from the DECLARE-LONG II (Drug-Eluting Stenting Followed
by Cilostazol Treatment Reduces Late Restenosis in Patients with Long Coronary Lesions) trial. J. Am. Coll. Cardiol. 2011, 57,
1264–1270.
107. Lee, Y.S.; Bae, H.J.; Kang, D.W.; Lee, S.H.; Yu, K.; Park, J.M.; Cho, Y.J.; Hong, K.S.; Kim, D.E.; Kwon, S.U.; et al. Cilostazol in
Acute Ischemic Stroke Treatment (CAIST Trial): A randomized double-blind non-inferiority trial. Cerebrovasc. Dis. 2011, 32, 65–71.
[CrossRef] [PubMed]
108. Suh, J.W.; Lee, S.P.; Park, K.W.; Lee, H.Y.; Kang, H.J.; Koo, B.K.; Cho, Y.S.; Youn, T.J.; Chae, I.H.; Choi, D.J.; et al. Multicenter
randomized trial evaluating the efficacy of cilostazol on ischemic vascular complications after drug-eluting stent implantation
for coronary heart disease: Results of the CILON-T (influence of CILostazol-based triple antiplatelet therapy ON ischemic
complication after drug-eluting stenT implantation) trial. J. Am. Coll. Cardiol. 2011, 57, 280–289. [PubMed]
109. Iida, O.; Yokoi, H.; Soga, Y.; Inoue, N.; Suzuki, K.; Yokoi, Y.; Kawasaki, D.; Zen, K.; Urasawa, K.; Shintani, Y.; et al. Cilostazol
reduces angiographic restenosis after endovascular therapy for femoropopliteal lesions in the Sufficient Treatment of Peripheral
Intervention by Cilostazol study. Circulation 2013, 127, 2307–2315. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 2593 25 of 26
110. Gao, W.; Zhang, Q.; Ge, H.; Guo, Y.; Zhou, Z. Efficacy and safety of triple antiplatelet therapy in obese patients undergoing stent
implantation. Angiology 2013, 64, 554–558. [CrossRef]
111. Youn, Y.J.; Lee, J.W.; Ahn, S.G.; Lee, S.H.; Choi, H.; Yu, C.W.; Hong, Y.J.; Kwon, H.M.; Hong, M.K.; Jang, Y.; et al. Multicenter
randomized trial of 3-month cilostazol use in addition to dual antiplatelet therapy after biolimus-eluting stent implantation for
long or multivessel coronary artery disease. Am. Heart J. 2014, 167, 241–248.e1. [CrossRef]
112. Uchiyama, S.; Sakai, N.; Toi, S.; Ezura, M.; Okada, Y.; Takagi, M.; Nagai, Y.; Matsubara, Y.; Minematsu, K.; Suzuki, N.; et al. Final
Results of Cilostazol-Aspirin Therapy against Recurrent Stroke with Intracranial Artery Stenosis (CATHARSIS). Cerebrovasc. Dis.
Extra 2015, 5, 1–13. [CrossRef] [PubMed]
113. Ueda, H.; Kido, A.; Matsuhisa, S.; Asawa, K.; Yoshida, N.; Tsujimoto, M.; Sasaki, Y.; Kuga, Y.; Yamasaki, M.; Ueda, K.; et al.
Addition of cilostazol to aspirin therapy for secondary prevention of cardiovascular and cerebrovascular disease in patients
undergoing percutaneous coronary intervention: A randomized, open-label trial. Am. Heart J. 2016, 173, 134–142. [CrossRef]
114. Zheng, X.T.; Chen, K.Y.; Liu, T.; Xu, L.X.; Che, J.J.; Rha, S.W.; Li, G.P. Low-dose adjunctive cilostazol in patients with complex
lesions undergoing percutaneous coronary intervention. Clin. Exp. Pharmacol. Physiol. 2016, 43, 29–33. [CrossRef]
115. Tang, Y.D.; Wang, W.; Yang, M.; Zhang, K.; Chen, J.; Qiao, S.; Yan, H.; Wu, Y.; Huang, X.; Xu, B.; et al. Randomized Comparisons
of Double-Dose Clopidogrel or Adjunctive Cilostazol Versus Standard Dual Antiplatelet in Patients with High Posttreatment
Platelet Reactivity: Results of the CREATIVE Trial. Circulation 2018, 137, 2231–2245. [CrossRef]
116. Lee, C.H.; Lee, J.Y.; Park, G.M.; Lee, S.W.; Kim, H.S.; Choi, Y.J.; Nam, C.W.; Cho, J.H.; Shin, W.Y.; Seo, J.B.; et al. Comparison of 1-
Year Outcomes of Triple (Aspirin + Clopidogrel + Cilostazol) Versus Dual Antiplatelet Therapy (Aspirin + Clopidogrel + Placebo)
after Implantation of Second-Generation Drug-Eluting Stents into One or More Coronary Arteries: From the DECREASE-PCI
Trial. Am. J. Cardiol. 2018, 121, 423–429.
117. Kim, B.J.; Lee, E.J.; Kwon, S.U.; Park, J.H.; Kim, Y.J.; Hong, K.S.; Wong, L.K.S.; Yu, S.; Hwang, Y.H.; Lee, J.S.; et al. Prevention of
cardiovascular events in Asian patients with ischaemic stroke at high risk of cerebral haemorrhage (PICASSO): A multicentre,
randomised controlled trial. Lancet Neurol. 2018, 17, 509–518. [CrossRef] [PubMed]
118. Hong, S.; Nam, M.; Little, B.B.; Paik, S.; Lee, K.; Woo, J.; Kim, D.; Kang, J.; Chun, M.; Park, Y. Randomized control trial comparing
the effect of cilostazol and aspirin on changes in carotid intima-medial thickness. Heart Vessel. 2019, 34, 1758–1768. [CrossRef]
[PubMed]
119. Toyoda, K.; Uchiyama, S.; Yamaguchi, T.; Easton, J.D.; Kimura, K.; Hoshino, H.; Sakai, N.; Okada, Y.; Tanaka, K.; Origasa, H.;
et al. Dual antiplatelet therapy using cilostazol for secondary prevention in patients with high-risk ischaemic stroke in Japan: A
multicentre, open-label, randomised controlled trial. Lancet Neurol. 2019, 18, 539–548. [CrossRef]
120. Chen, Y.C.; Lin, F.Y.; Lin, Y.W.; Cheng, S.M.; Lin, R.H.; Chuang, C.L.; Sheu, J.S.; Chen, S.M.; Chang, C.C.; Tsai, C.S. DAPT Plus
Cilostazol is Better Than Traditional DAPT or Aspirin Plus Ticagrelor as Elective PCI for Intermediate-to-Highly Complex Cases:
Prospective, Randomized, PRU-Based Study in Taiwan. Am. J. Cardiovasc. Drugs 2019, 19, 75–86. [CrossRef]
121. Uchiyama, S.; Toyoda, K.; Omae, K.; Saita, R.; Kimura, K.; Hoshino, H.; Sakai, N.; Okada, Y.; Tanaka, K.; Origasa, H.; et al. Dual
Antiplatelet Therapy Using Cilostazol in Patients with Stroke and Intracranial Arterial Stenosis. J. Am. Heart Assoc. 2021, 10,
e022575. [CrossRef]
122. Kalantzi, K.; Tentolouris, N.; Melidonis, A.J.; Papadaki, S.; Peroulis, M.; Amantos, K.A.; Andreopoulos, G.; Bellos, G.I.; Boutel, D.;
Bristianou, M.; et al. Efficacy and Safety of Adjunctive Cilostazol to Clopidogrel-Treated Diabetic Patients with Symptomatic
Lower Extremity Artery Disease in the Prevention of Ischemic Vascular Events. J. Am. Heart Assoc. 2021, 10, e018184. [CrossRef]
123. Sohn, M.; Chun, E.J.; Lim, S. Cilostazol treatment for preventing adverse cardiovascular events in patients with type 2 diabetes
and coronary atherosclerosis: Long-term follow-up of the ESCAPE study. J. Diabetes 2022, 14, 524–531. [CrossRef] [PubMed]
124. Lin, J.L.; Tseng, W.K.; Lee, P.T.; Lee, C.H.; Tseng, S.Y.; Chen, P.W.; Chang, H.Y.; Chao, T.H. A Randomized Controlled Trial
Evaluating Outcome Impact of Cilostazol in Patients with Coronary Artery Disease or at a High Risk of Cardiovascular Disease. J.
Pers. Med. 2022, 12, 938. [CrossRef] [PubMed]
125. Wardlaw, J.M.; Woodhouse, L.J.; Mhlanga, I.I.; Oatey, K.; Heye, A.K.; Bamford, J.; Cvoro, V.; Doubal, F.N.; England, T.; Hassan, A.;
et al. Isosorbide Mononitrate and Cilostazol Treatment in Patients with Symptomatic Cerebral Small Vessel Disease: The Lacunar
Intervention Trial-2 (LACI-2) Randomized Clinical Trial. JAMA Neurol. 2023, 80, 682–692. [CrossRef]
126. McHutchison, C.; Blair, G.W.; Appleton, J.P.; Chappell, F.M.; Doubal, F.; Bath, P.M.; Wardlaw, J.M. Cilostazol for Secondary
Prevention of Stroke and Cognitive Decline: Systematic Review and Meta-Analysis. Stroke 2020, 51, 2374–2385. [CrossRef]
[PubMed]
127. Muhammad, I.F.; Borne, Y.; Zaigham, S.; Soderholm, M.; Johnson, L.; Persson, M.; Melander, O.; Engstrom, G. Comparison of risk
factors for ischemic stroke and coronary events in a population-based cohort. BMC Cardiovasc. Disord. 2021, 21, 536. [CrossRef]
128. Xie, W.; Zheng, F.; Zhong, B.; Song, X. Long-Term Antiplatelet Mono- and Dual Therapies after Ischemic Stroke or Transient
Ischemic Attack: Network Meta-Analysis. J. Am. Heart Assoc. 2015, 4, e002259. [CrossRef] [PubMed]
129. Park, S.; Rha, S.W.; Choi, B.G.; Kim, W.; Choi, W.G.; Lee, S.J.; Lee, J.B.; Park, J.Y.; Park, S.M.; Jeong, M.H.; et al. Efficacy and safety
of cilostazol-based triple antiplatelet therapy compared with clopidogrel-based dual antiplatelet therapy in patients with acute
ST-elevation myocardial infarction undergoing percutaneous coronary intervention: A multicenter, randomized, open-label,
phase 4 trial. Am. Heart J. 2023, 265, 11–21.
130. Kherallah, R.Y.; Khawaja, M.; Olson, M.; Angiolillo, D.; Birnbaum, Y. Cilostazol: A Review of Basic Mechanisms and Clinical
Uses. Cardiovasc. Drugs Ther. 2022, 36, 777–792. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 2593 26 of 26
131. Brown, T.; Forster, R.B.; Cleanthis, M.; Mikhailidis, D.P.; Stansby, G.; Stewart, M. Cilostazol for intermittent claudication. Cochrane
Database Syst. Rev. 2021, 6, CD003748. [CrossRef]
132. Dawson, D.L.; Cutler, B.S.; Hiatt, W.R.; Hobson, R.W., 2nd; Martin, J.D.; Bortey, E.B.; Forbes, W.P.; Strandness, D.E., Jr. A
comparison of cilostazol and pentoxifylline for treating intermittent claudication. Am. J. Med. 2000, 109, 523–530. [CrossRef]
133. Strandness, D.E., Jr.; Dalman, R.L.; Panian, S.; Rendell, M.S.; Comp, P.C.; Zhang, P.; Forbes, W.P. Effect of cilostazol in patients
with intermittent claudication: A randomized, double-blind, placebo-controlled study. Vasc. Endovasc. Surg. 2002, 36, 83–91.
[CrossRef] [PubMed]
134. de Franciscis, S.; Gallelli, L.; Battaglia, L.; Molinari, V.; Montemurro, R.; Stillitano, D.M.; Buffone, G.; Serra, R. Cilostazol prevents
foot ulcers in diabetic patients with peripheral vascular disease. Int. Wound J. 2015, 12, 250–253. [CrossRef] [PubMed]
135. Gerhard-Herman, M.D.; Gornik, H.L.; Barrett, C.; Barshes, N.R.; Corriere, M.A.; Drachman, D.E.; Fleisher, L.A.; Fowkes, F.G.R.;
Hamburg, N.M.; Kinlay, S.; et al. 2016 AHA/ACC Guideline on the Management of Patients with Lower Extremity Peripheral
Artery Disease: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force
on Clinical Practice Guidelines. J. Am. Coll. Cardiol. 2017, 69, 1465–1508. [CrossRef]
136. Senbokuya, N.; Kinouchi, H.; Kanemaru, K.; Ohashi, Y.; Fukamachi, A.; Yagi, S.; Shimizu, T.; Furuya, K.; Uchida, M.; Takeuchi,
N.; et al. Effects of cilostazol on cerebral vasospasm after aneurysmal subarachnoid hemorrhage: A multicenter prospective,
randomized, open-label blinded end point trial. J. Neurosurg. 2013, 118, 121–130. [CrossRef]
137. Takagi, T.; Hara, H. Protective effects of cilostazol against hemorrhagic stroke: Current and future perspectives. J. Pharmacol. Sci.
2016, 131, 155–161. [CrossRef]
138. Lee, J.H.; Park, S.Y.; Shin, H.K.; Kim, C.D.; Lee, W.S.; Hong, K.W. Protective effects of cilostazol against transient focal cerebral
ischemia and chronic cerebral hypoperfusion injury. CNS Neurosci. Ther. 2008, 14, 143–152. [CrossRef] [PubMed]
139. Lim, J.H.; Woo, J.S.; Shin, Y.W. Cilostazol protects endothelial cells against lipopolysaccharide-induced apoptosis through ERK1/2-
and P38 MAPK-dependent pathways. Korean J. Intern. Med. 2009, 24, 113–122. [CrossRef] [PubMed]
140. Watanabe, T.; Zhang, N.; Liu, M.; Tanaka, R.; Mizuno, Y.; Urabe, T. Cilostazol protects against brain white matter damage and
cognitive impairment in a rat model of chronic cerebral hypoperfusion. Stroke 2006, 37, 1539–1545. [CrossRef]
141. Takagi, T.; Imai, T.; Mishiro, K.; Ishisaka, M.; Tsujimoto, M.; Ito, H.; Nagashima, K.; Matsukawa, H.; Tsuruma, K.; Shimazawa, M.;
et al. Cilostazol ameliorates collagenase-induced cerebral hemorrhage by protecting the blood-brain barrier. J. Cereb. Blood Flow.
Metab. 2017, 37, 123–139. [CrossRef]
142. Lee, J.H.; Park, S.Y.; Shin, Y.W.; Hong, K.W.; Kim, C.D.; Sung, S.M.; Kim, K.Y.; Lee, W.S. Neuroprotection by cilostazol, a
phosphodiesterase type 3 inhibitor, against apoptotic white matter changes in rat after chronic cerebral hypoperfusion. Brain Res.
2006, 1082, 182–191. [CrossRef] [PubMed]
143. Shichinohe, H.; Tan, C.; Abumiya, T.; Nakayama, N.; Kazumata, K.; Hokari, M.; Houkin, K.; Kuroda, S. Neuroprotective effects of
cilostazol are mediated by multiple mechanisms in a mouse model of permanent focal ischemia. Brain Res. 2015, 1602, 53–61.
[CrossRef] [PubMed]
144. Kim, B.C.; Youn, Y.C.; Jeong, J.H.; Han, H.J.; Kim, J.H.; Lee, J.H.; Park, K.H.; Park, K.W.; Kim, E.J.; Oh, M.S.; et al. Cilostazol Versus
Aspirin on White Matter Changes in Cerebral Small Vessel Disease: A Randomized Controlled Trial. Stroke 2022, 53, 698–709.
[CrossRef] [PubMed]
145. Pratt, C.M. Analysis of the cilostazol safety database. Am. J. Cardiol. 2001, 87, 28D–33D. [CrossRef]
146. Lee, D.H.; Chun, E.J.; Moon, J.H.; Yun, H.M.; Lim, S. Effect of cilostazol on carotid plaque volume measured by three-dimensional
ultrasonography in patients with type 2 diabetes: The FANCY study. Diabetes Obes. Metab. 2020, 22, 2257–2266. [CrossRef]
147. Otsuka. Pletal—Highlights of Prescribing Information; Otsuka: Tokyo, Japan, 1999.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.