You are on page 1of 11

114 Send Orders for Reprints to reprints@benthamscience.

ae

Current Vascular Pharmacology, 2018, 16, 114-124


REVIEW ARTICLE
ISSN: 1570-1611
eISSN: 1875-6212

Current Vascular
Pharmacology
The journal for current and in-depth reviews on Vascular Pharmacology

Pharmacological Therapy of Abdominal Aortic Aneurysm: An Update Impact


Factor:
2.391

BENTHAM
SCIENCE

Yi-dong Wang1, Zhen-jie Liu2, Jun Ren3 and Mei-xiang Xiang1,*

1
Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Cardiovascular Key
Lab of Zhejiang Province, #88 Jiefang Road, Hangzhou, Zhejiang, 310009, P.R. China; 2Department of Vascular Sur-
gery, Second Affiliated Hospital of Zhejiang University School of Medicine, #88 Jiefang Road, Hangzhou, Zhejiang,
310009, P.R. China; 3Department of Surgery, University of Wisconsin - Madison, Madison, WI, USA

Abstract: Background: Abdominal aortic aneurysm (AAA), a progressive segmental abdominal aortic
dilation, is associated with high mortality. AAA is characterized by inflammation, smooth muscle cell
ARTICLE HISTORY (SMC) depletion and extracellular matrix (ECM) degradation. Surgical intervention and endovascular
therapy are recommended to prevent rupture of large AAAs. Unfortunately, there is no reliable pharmacol-
Received: February 03, 2017
Revised: March 24 2017 ogical agent available to limit AAA expansion. In the past decades, extensive investigations and a body of
Accepted: March 26, 2017
ongoing clinical trials aimed at defining potent treatments to inhibit and even regress AAA growth.
DOI:
10.2174/1570161115666170413145705 Conclusion: In this review, we summarized recent progress of potential strategies, particularly macrol-
ides, tetracyclines, statins, angiotensin converting enzyme inhibitors, angiotensin receptor blocker, cor-
ticosteroid, anti-platelet drugs and mast cell stabilizers. We also consider recently identified novel mo-
lecular targets, which have potential to be translated into clinical practice in the future.
Current Vascular Pharmacology

Keywords: Abdominal aortic aneurysm, clinical trials, medical treatment, pharmacological therapy, molecular target, disease.

1. INTRODUCTION 2. CURRENT MEDICAL TREATMENTS FOR AAA


Abdominal aortic aneurysm (AAA), a progressive ab- 2.1. Macrolides and Tetracyclines
dominal aortic dilation due to various life-style and genetic
Macrolides, roxithromycin and azithromycin, can
factors, is associated with high mortality. The incidence of
potently combat various infections including Chlamydia
AAA is up to 8% in man over 65 years old [1, 2]. In the
pneumonia (C. pneumonia). It has been reported that C.
United States, AAA rupture and complications after sur-
gery caused at least 15,000 deaths/year, and the actual pneumonia is associated with AAA formation [3]. Thus, an-
tichlamydial treatment is a putative way to control AAA.
number of deaths may double considering those without
Administration of roxithromycin 300 mg daily for 4 weeks
diagnosis [2].
significantly decreased AAA expansion compared with pla-
Although significant progress has been achieved in cebo treatment in the 1.5 year follow-up [3]. Furthermore, a
AAA imaging as well as in open and endovascular repair, long-term effects of roxithromycin were evaluated [4]. Pa-
effective pharmacological treatments (i.e. for the preven- tients were treated with roxithromycin 300 mg daily for 4
tion of AAA occurrence, and expansion, rupture as well as weeks annually and followed up for average 5.27 years.
perioperative treatment of AAA) are still unavailable Consistently, treatment of roxithromycin reduced annual
(Table 1). Current evidence of medical therapy is low- growth rate, namely 36%, and lowered the risk of surgical
quality, and no effective and specific target has been found repair [4]. Nevertheless, a randomized double-blind con-
for AAA, likely due to incomplete understanding of the trolled trial demonstrated that azithromycin hardly inter-
biological mechanisms underlying the disease. Therefore, vened AAA expansion, suggesting a limited role of macrol-
surgical intervention is the optimal treatment for patients ides in AAA treatment [5]. Elimination of C. pneumonia
with AAA. Herein, we review the most up-to-date pharma- seems not to fully account for the effectiveness of roxithro-
cological treatments for AAA and consider some potential mycin. The underlying mechanisms needs to be explored in
target molecules. the future.
The role of doxycycline in AAA was extensively dis-
cussed in a recent review [6]. In elastase- or angiotensin
*Address correspondence to this author at the Department of Cardiology, (Ang) II-induced AAA models, doxycycline strongly inhib-
Second Affiliated Hospital of Zhejiang University School of Medicine, ited AAA formation, but it had little effect on established
Hangzhou, China; Tel: +86-13588035801; E-mail: xiangmxhz@163.com AAAs [6, 7]. The effects of doxycycline on AAA were origi-

1875-6212/18 $58.00+.00 © 2018 Bentham Science Publishers


Pharmacological Therapy of Abdominal Aortic Aneurysm Current Vascular Pharmacology, 2018, Vol. 16, No. 2 115

Table 1. Recent randomized clinical trials of AAA medical treatments.

Medication Target Intervention Primary Outcome Trial Registration Status

ACZ885 IL-1 Subcutaneous ACZ885 (monthly) vs placebo AAA growth by ultrasound NCT02007252 Terminated

Telmisartan RAS system Telmisartan (40 mg daily) vs placebo AAA growth by CTA NCT01683084 Active

Perindopril Perindopril arginine (10 mg daily) vs amlodip-


RAS system AAA growth by ultrasound NCT01118520 Completed
arginine ine (5 mg daily) vs placebo

Ticagrelor Platelet Ticagrelor (90 mg daily) vs placebo AAA growth by MRI NCT02070653 Recruiting

Allogeneic T-regulatory 1 million allogeneic MSCs/kg vs 3 million Incidence of treatment Not yet
NCT02843854
MSCs cells allogeneic MSCs/kg vs placebo (Plasmalyte A) related adverse events recruiting

Doxycycline MMPs Doxycycline (100 mg twice daily) vs placebo AAA growth by CT NCT01756833 Recruiting

-adrenergic
Atenolol and Atenolol (50 mg daily) vs valsartan
receptor and AAA growth by CT NCT01904981 Recruiting
Valsartan (80 mg daily)
RAS system

CRD007 (10 mg twice daily) vs CRD007 (25


CRD007 Mast cell mg twice daily) vs CRD007 (40 mg twice AAA growth by ultrasound NCT01354184 Completed
daily) vs placebo

Aliskiren and RAS system and Aliskiren (150 mg daily) vs amlodipine Aneurysmal vessel wall
NCT01425242 Terminated
amlodipine calcium channel (5 mg daily) inflammation by PET-CT

originally attributed to its effect on matrix metalloproteinases A body of observational clinical studies indicated that
(MMPs) activity reduction by competitive binding zinc co- AAA patients on statins had a lower growth rate, compared
factor. New insight theory proposed that doxycycline possi- with non-statin users [17-20]. Nevertheless, another four in-
bly decreased T cell expansion during AAA growth [6]. Sev- vestigations, as Dunne concluded, failed to detect a difference
eral small clinical trials supported the inhibitory effects of in AAA growth between statin users and non-users [17].
doxycycline on AAA. However, a recent large, randomized, These discrepancies might stem from different AAA meas-
placebo-controlled, double-blind trial challenged this idea [6- urements. Although a variety of meta-analyses of this topic
8]; 286 patients who were treated with 100 mg doxycycline suggested that statin therapy was related to less AAA dilation,
daily for 18 months did not gain benefit during AAA pro- this conclusion relied on low-quality evidence [21-25]. To
gression. However, they had a higher AAA expansion rate. confirm the roles of statin in limiting AAA expansion, a large
The insufficient dose, 100 mg daily, may responsible for the randomized trial is required in the future. Regardless of effects
negative result. Another phase IIb, randomized, double-blind of statins on AAA expansion, several early studies claimed
trial with 200 mg daily is still on-going, and their newly pub- that these drugs effectively improved the outcome of patients
lished paper indicated that data advocates the hypothesis that with vascular surgery [26-28]. In a nationwide, case-control
AAA development declined by 40% [9]. The ultimate con- study, statins significantly lowered 30-day mortality rate of
clusion of this trial will potentially provide solid evidence for ruptured AAA patients [29]. Promisingly, continuous statin
clinical practice. treatment during the perioperative and postoperative period in
the long run increased survival after open or endovascular
2.2. Statins aneurysm repair (EVAR) and reduced incidence of complica-
tions [30-32]. Following up patients after surgery for 6 years,
Emerging evidence suggested that statins are promising
de Bruin's work revealed that statin use is independently asso-
candidates in AAA medical therapy. Several studies in mur-
ciated with better outcome after AAA repair [32]. It has also
ine models reported that statin administration decreased
been reported that statin treatment intended to regress the
AAA growth, but Iida et al. declared a negative outcome [10,
AAA sac after EVAR, though no significant difference was
11]. The influence of statins on AAA may be multifactorial observed compared to the group without statin [33, 34]. There-
involving suppression of inflammation and ECM remodeling
fore, the available evidence suggests that statins should be
in addition to their lipid-lowering capability [12, 13]. In hu-
recommended for AAA patients.
man AAA specimens, administration of simvastatin or
atorvastatin greatly depressed vascular wall inflammation,
2.3. Angiotensin Converting Enzyme Inhibitors (ACEI)
reflected by decreased nuclear factor-B (NF-B) activity
and Angiotensin Receptor Blockers (ARB)
and its downstream cytokines, interleukin (IL)-6 and mono-
cyte chemotactic protein (MCP)-1 [14, 15]. Furthermore, a The renin-angiotensin system (RAS) plays a role in the
recent study suggested that simvastatin treatment also regu- pathophysiological process of AAA [35]. The molecular
lated MMPs and their inhibitors, namely tissue inhibitor of mechanisms involved in AAA formation are elaborated in
metalloproteinases (TIMPs) in human AAA tissues and several reviews, suggesting that elevation of MCP-1, cy-
thrombi [16]. clooxygenase-2, osteopontin, oxidative stress and MMPs me-
116 Current Vascular Pharmacology, 2018, Vol. 16, No. 2 Wang et al.

Table 2. Clinical studies on the effect of ACEI and ARB on AAA.

Year No. of Patients Design Follow-up (Years) Effect Reference

2002 438 cohort 4 ACEI did not affect AAA growth rate [147]

2006 15326 case-control NA ACEI prevent AAA rupture [36]

2010 1269 cohort 3.4 ARB decreased AAA growth rate [37]

2010 883 cohort NA ACEI or ARB increased perioperative mortality of AAA repair [42]

2010 1701 cohort 1.9 ACEI enhanced AAA growth [41]

2010 652 cohort 5 ACEI did not relate to AAA growth [148]

2014 122 cohort 0.67 ACEI and ARB attenuated AAA expansion [38]

2015 9441 cohort 5 ACEI and ARB reduced AAA mortality, ACEI reduced risk of surgery [39]

2016 3586 case-control NA ACEI and ARB were not associated with AAA rupture [40]

2016 224 RCT over 2 ACEI did not affect AAA growth rate [43]

No. = number; RCT = randomized controlled trial; NA = not applicable.

diated the effects of Ang II on the aortic wall [12, 35]. A body AAA (IAAA), which is characterized by thickening wall in
of clinical investigations have been accumulated in past 10 aneurysmal portion and fibrous adhesion to adjacent tissue
years, but their results are divergent (Table 2). A retrospective [45-47]. Corticosteroids mitigated the fibrosis and if there is
case-control study of patients with rupture or intact AAA pre- no urgency to operate (diameter <5.5 cm), it is the optimal
sented that ACEI, rather than ARB, effectively reduced risk of choice in IAAA [45]. However, until now no guidelines of
AAA rupture, but the fact that 1% patients received ARB in medical treatment for IAAA exist. Although an observa-
this study weakened the conclusion [36]. Nevertheless, a pro- tional study denied the relationship between corticosteroid
gram lasting 25 years of surveillance pointed out that ARB and AAA growth rate, a recent randomized, double-blind,
seems to reduce AAA expansion although the effects were not placebo-controlled clinical trial concluded that preoperative
independent of all confounders [37]. Recently, two observa- administration of methylprednisolone facilitated recovery
tional investigations validated the role of ACEI and ARB in after endovascular AAA repair [48]. Methylprednisolone 30
inhibition of AAA [38, 39]. One study collected 9441 patients mg/kg body weight alleviated the systemic inflammatory
from nation-wide registries; ACEI and ARB comparably de- response syndrome (SIRS) from 92-27%, accompanied by
creased mortality, and ACEI users also had a lower risk of the reduction of interleukin-6 and C-reactive protein [48].
surgery for AAA repair [39]. In contrast, another case-control
follow-up study demonstrated no clear relationship between 2.5. Anti-platelet Drugs
RAS blockade and risk of rupture of AAA but lack of smok-
Intraluminal mural thrombus (ILT) includes inflamma-
ing information may limit credibility [40]. What is worse,
Sweeting and Railton’s observational trial claimed that the tory cells, particularly neutrophils, pro-inflammatory cytoki-
nes and proteolytic enzymes, and plays a central role in AAA
administration of ACEI increased AAA growth rate and pe-
progression and rupture [49]. In a small prospective cohort,
rioperative mortality [41, 42]. These inconsistencies derived
the volume of infrarenal aorta thrombus measured by com-
from different imaging measurements, number of patients,
puted tomography angiography (CTA) in AAA patients was
baseline characteristics and statistical methodology. Recently,
positively associated with the adverse cardiovascular events
the result of AARDVARK trail, the first randomized trial to
evaluate the effects of ACEI on AAA, was released [43]. 224 and AAA growth rate [50]. In Ang II-induced AAA models,
anti-platelet drugs significantly diminished AAA formation,
patients were randomized to three groups who were treated
and intriguingly, greatly ameliorated rupture-induced death
with perindopril arginine 10 mg, amlodipine 5 mg or placebo,
in established AAAs [51, 52]. These effects were accompa-
respectively and followed more than 2 years. Administration
nied by a reduction in macrophage aggregation and MMP
of perindopril failed to significantly prevent small AAA dila-
expression [51]. Currently, AAA patients are recommended
tion annually despite the fact that blood pressure was lower in
this group [43]. The number of patients with AAA diameters to receive low-dose aspirin, but compelling evidence is still
lacking [53, 54]. In agreement with animal experiments,
that reached 5.5 cm or who underwent surgery were also simi-
some observational studies demonstrated that anti-platelet
lar among these groups. It is suggested that ACEI is not an
treatment decelerated medium-sized AAA expansion and
effective choice for AAA treatment. Another randomized
lowered dissection or rupture rate [51, 55]. Conversely, oth-
clinical trial of ARB is still on-going [44].
ers contradicted these views [22, 37, 56]. Predominantly,
2.4. Corticosteroids Chen’s national retrospective cohort study suggested that
low dose aspirin did not protect AAA patients from mortality
Several case reports demonstrated that corticosteroid or adverse events after 10-year follow-up [56]. Also, pread-
therapy was effective for the patients with inflammatory mission use of low dose aspirin did not change the risk of
Pharmacological Therapy of Abdominal Aortic Aneurysm Current Vascular Pharmacology, 2018, Vol. 16, No. 2 117

AAA rupture in hospital and even worsened 30-day mortal- macrophage infiltration [68, 69]. Similarly, analysis of
ity [57]. Collectively, no high-quality randomized controlled specimens obtained from aneurysmectomy demonstrated that
trials evaluated the importance anti-platelet agents for AAA 15 mg/day of PPAR- agonist for >2 months decreased
patients. A randomized double-blind controlled trial of tica- macrophage infiltration, TNF- and MMP-9 in the aortic
grelor, a new anti-platelet drug, is in progress. This trial is wall and periaortic fat [70]. The effects of PPAR- agonists
expected to provide evidence about the role of anti-platelet on aorta are pleiotropic (Fig. 1). PPAR- deficiency in SMCs
drugs in AAA [58]. accelerated AAA development and elastin disintegration,
possibly by increasing cathepsin S [67, 71]. Additionally,
2.6. Mast Cell Stabilizers inhibition of PPAR- in aortic adventitial fibroblasts inter-
fered synthesis of elastic fibre components [67]. Also, spe-
Mast cells play vital roles in allergic responses, tumour
cific delivery of PPAR- agonists to macrophage drove
and atherosclerosis and studies also emphasized their signifi- macrophage towards the M2 type, which has anti-
cance in AAA pathogenesis [59]. Mast cell deficiency or its
inflammatory effects [72]. These studies suggested PPAR-
stabilizers retarded the AAA progression, while activation of
agonists potentially restricted AAA progression, but there is
mast cell aggravated AAA growth [60, 61]. The mechanistic
a need for further validation.
analysis showed that mast cells sparked angiogenesis, SMC
apoptosis, and elevation of MMPs. Proinflammatory factors The AKT signaling pathway, which can be induced by
including IL-6 and IFN-, chymase, and tryptase mediated growth factors and cytokines, is involved in cellular activi-
these effects [60, 62]. The abovementioned evidence sug- ties such as metabolism, proliferation and migration. Phos-
gested that mast cell stabilizers, which are extensively used phorylated AKT and total AKT were elevated in both AAA
clinically to combat allergic disease, may also become a mouse models and human AAAs. LY294002, an AKT in-
candidate for AAA treatment. Disappointedly, the AORTA hibitor, attenuated the expansion of AAA [73]. In vitro ex-
trial, which explored the relationship between mast cell in- periments claimed that macrophages and SMCs treated with
hibitor, pemirolast, and medium-sized AAA expansion, re- elastase had increased AKT phosphorylation, and MMP9
vealed a negative conclusion [63]; 326 patients treated with expression, and these effects were abandoned by the siRNA
10, 25, 40 mg pemirolast or placebo for 1 year had a similar against AKT [73]. However, another study indicated that a
AAA growth rate. The insufficient dose and study interval lack of AKT2 deteriorated Ang II-induced AAA. The pres-
may account for this result. Additionally, medium-sized ence of AKT2 reduced MMP9 and fostered TIMP-1 in hu-
AAA, namely 39-49 mm in diameter, is a late stage for ef- man aortic SMC [74]. Furthermore, fenofibrate inhibited
fectiveness of mast cell stabilizers, which can be assessed in aortic dilatation and atheroma progression partially because
animal models. Varying dosages and long study intervals are of up-regulated AKT1 and eNOS [75]. The individual effects
awaited in the future. of AKT isoforms are possibly not the same as total AKT.
Different models are also a potential explanation for these
3. NEW THERAPEUTIC TARGETS FOR MEDICAL disparities.
TREATMENTS OF AAA Prostaglandin E2 (PGE2) is involved in multiple vascu-
3.1. Several Critical Signaling Pathways lar inflammatory process including atherosclerosis and AAA,
and EP1-4 receptors mediated its effects [76]. Although EP-
Chronic inflammation has long been recognized as a 2, -3, -4 are detected in mouse and human AAA tissues, EP-
crucial pathological event in AAAs. NF-B and ets, two 4 is the only receptor which plays a role in AAA develop-
regulators of inflammation, have been reported to be associ-
ment [77]. EP-4 knockout or systemic administration of EP-
ated with AAA expansion [64, 65]. In AAA samples ob-
4 antagonist strikingly alleviated AAA dilation, and IL-6, IL-
tained from humans, NF-B and ets were observed to be
17, MIP-1MMP-2 and MMP-9 are concomitantly reduced
greatly activated [64, 65]. Perivascular delivery of chimeric
[78, 79]. Another mechanistic study asserted that EP-4 was
decoy oligodeoxynucleotides (ODNs) efficiently repressed
NF-B and its expression and subsequently slowed AAA highly expressed in the microvasculature of AAA specimens,
dilation in rat and rabbit models [64, 66]. Predominantly, and activation of EP-4 in microvascular endothelial cells
ODNs are able to reduce aortic diameter in an established induced angiogenesis in vitro [80]. It has been indicated that
rabbit AAA model [65]. Exploration of mechanisms sug- EP-4 was potentially implicated in AAA-related vasculariza-
gested inhibition of NF-B and ets significantly decreased tion. These stimulating results predicted that EP-4 antagonist
MMPs production and macrophage infiltration, and facili- is an alternative target for AAA treatment. The only uncer-
tated elastin and collagen synthesis, which was vital to aortic tainty stemmed from Tang’s work that suggested that a lack
remodelling [65]. These results suggested that ODNs were of EP-4 in bone marrow-derived cells exacerbated AAA for-
potential therapy for AAA management, but the delivery mation [81]. Thus, macrophage specific inhibition of EP-4
method should be further developed may not be a rational choice for therapy.
Peroxisome proliferator-activated receptor (PPAR)- is Notch signaling pathway, which is composed of 4 Notch
ubiquitously expressed in all aortic cells including SMCs, receptors 1-4, is highly conserved and ubiquitously ex-
inflammatory cells and fibroblasts. The expression PPAR- pressed in various species. A series of recent studies linked
is higher in AAA samples from humans and mice [67]. Ad- Notch signaling to AAA formation, and Notch1 particularly
ministration of PPAR- agonists, rosiglitazone or pioglita- plays a vital role [82-84]. In AAA patient’s plasma, Notch1
zone, which are used for type 2 diabetes mellitus treatment, level was increased compared with controls [85]. Further,
profoundly reduced aortic dilatation and rupture rate accom- Notch1 signaling was also enhanced in Ang II-induced mice
panied by a decline of proinflammatory cytokines and and human AAA tissues, while Doyle reported that Notch1
118 Current Vascular Pharmacology, 2018, Vol. 16, No. 2 Wang et al.

6PRRWKPXVFOHFHOOV &DWKHSVLQ6

33$5ȖDJRQLVW

)LEUREODVWV (ODVWLQDQG)LEXOLQ

0 0

0DFURSKDJHV
Fig. (1). The pleiotropic effects of PPAR- agonists on AAA.

was reduced in tissues obtained from human detected by intravenously via tail vein in Ang II-induced AAA model,
PCR [82, 84, 86]. It is not clear how this difference occurred. and consistently retarded AAA development [94]. Encourag-
Using Notch1 haploinsufficiency mice, Hans’ group demon- ingly, delivery of MSCs in a catheter-dependent way stabi-
strated that Notch1 deficiency reduced AAA progression lized and even regressed the formed AAA, similar to clinical
[82]. This protective effect may be resulted from regulation patients [95, 96]. The effects of MSCs on AAA are plei-
of macrophage polarization, namely, less M1 macrophage otropic, and reduction of MMP-2, MMP-9, IL-6, MCP-1 and
and more M2 macrophage. In parallel, administration of elevation of TIMP-1 may be involved. Notably, Sharma’s
Notch1 inhibitor also had similar protection against AAA, studies demonstrated that MSCs mitigated HMGB1 expres-
implying Notch1 was a potential target for AAA therapy [83, sion in nicotinamide adenine dinucleotide phosphate oxidase
84]. The clinical use of these inhibitors should be evaluated (Nox2)-dependent manner in macrophage, and subsequently
in future investigations. inhibited production of IL-17 by CD4+ T cells [97, 98].
Overall, MSCs are promising therapy to alleviate AAA,
Toll-Like Receptors (TLRs) play pivotal roles in innate
though several factors such as delivery method and cell
and adaptive immune response, and are also implicated in
AAA development. Knockout of TLR4 reduced AAA dila- number need to be further evaluated. Also, the efficacy of
MSCs from different origins such as bone marrow and pla-
tion in Ang II and CaCl2-induced murine models [87, 88].
centa should be assessed, since these kinds of MSCs are ap-
Mechanistic studies reported that loss of TLR4 in SMC de-
plied in past studies [93, 98].
creased expression of IL-6 and MCP-1, but hardly affected
macrophages [87, 88]. On the other hand, Qin and his co- Additionally, adoptive transfer of regulatory T cells
workers reported that STAT3 phosphorylation was also a (Tregs) and M2 macrophages are reported to effectively al-
downstream pathway of activated TLR4, and TLR4 defi- leviated AAA development. Intravenous injection of Tregs
ciency undermined Ang II-induced STAT3 phosphorylation significantly reduced AAA expansion in Ang II-induced
[89]. Likewise, inhibitors of TLR4 such as Eritoran and AAA model, and concomitantly lowered MMPs and proin-
IAXO-102, effectively diminished AAA development, sug- flammatory cytokines [99, 100]. Conversely, Tregs origi-
gesting TLR4 was a potential target for AAA treatment [89, nated from IL-10-/- mice failed to inhibit AAA dilation, sug-
90]. In contrast, the role of TLR2 in AAA is still undeter- gesting that IL-10 mediated the beneficial effects of Tregs
mined. Loss of TLR2 hardly influenced AAA pathogenesis, [99, 100]. M2 macrophages, known as anti-inflammatory
but Yan’s suggested antagonists of TLR2 strikingly de- phenotype, were transferred into CaCl2-induced AAA model
creased AAA, accompanied by the reduction of inflamma- and dramatically decreased aortic diameter expansion [101].
tory cytokines and MMPs [87, 91]. The involvement of Tregs and M2 macrophages are two candidates for AAA
TLR2 in AAA is complex, and further studies are necessary treatment.
to assess therapeutic effects.
3.3. Kinases
3.2. Cell Therapy
c-Jun N terminal kinase (c-JNK), one member of mito-
Mesenchymal stem cells (MSCs) not only are able to gen-activated protein kinase (MAPK), is also involved in
differentiate into various cell types, but also show potent AAA formation [102, 103]. In Yoshimura’s encouraging
anti-inflammatory and immunoregulatory properties [92]. study, phosphorylated c-JNK was greatly increased, which
Previously, implantation of bone marrow-derived MSC enhanced MMPs activities and deteriorated tissue repair by
(BM-MSC) on aortic adventitia significantly decreased AAA reduction collagen and elastin [15, 102]. In vivo administra-
expansion, and inflammatory cytokines and MMPs were tion of SP600123, a JNK inhibitor, was observed to not only
reduced [93]. To be less invasive, BM-MSCs were injected limit AAA development, but also lead to regression of estab-
Pharmacological Therapy of Abdominal Aortic Aneurysm Current Vascular Pharmacology, 2018, Vol. 16, No. 2 119

lished AAAs in CaCl2 and Ang II-induced model through els, but whether these targets can be translated into clinical
augment of ECM synthesis [102]. In wild type mice treated practice remains unclear. Systemic injection of agonists or
with Ang II and nicotine, Guo demonstrated that administra- antagonists inevitably results in severe side effects and local
tion of SP600123 abrogated AAA formation [104]. These administration possibly minimized their effects [115].
promising results point to the need for clinical trials.
3.5. Cytokines
Another member of MAPKs, extracellular signal-related
kinase (ERK), was enhanced in elastase-induced AAA Interleukins (IL) are vital mediators of inflammatory
model and human samples, reflected by protein level of reactions and supposed to be associated with AAA develop-
phosphorylated ERK [105]. Mice without ERK-1 rarely de- ment. In human and elastase-induced murine models, IL-1
veloped AAA and MMP2 was predominately reduced in the was significantly elevated, and knockout of IL-1 and IL-1
meantime, suggesting that ERK-1 possibly regulated MMP2 receptor decreased AAA dilation, concomitantly reduced
production [105]. In contrast, analysis of specimens har- macrophage and neutrophil infiltration and maintained
vested from humans elucidated that ruptured AAAs con- elastin integrity [116]. Anakinra, an antagonist of IL-1 recep-
tained high level of MMP2 but low level of ERK-1, chal- tor, also disrupted AAA progression in a dose-dependent
lenging the relationship between ERK and MMP2 [106]. manner, suggesting it is a potential treatment for AAA [116].
ERK inhibitors can be applied in AAA models in mice in the IL-6 is abundant in AAA tissues, and circulating IL-6 may
future [107]. function as a biomarker for AAA [117, 118]. Disappoint-
Protein kinase C-delta (PKC-), a serine/threonine ingly, lack or inhibition of IL-6 declined thoracic aortic an-
kinase, has been reported to mediate SMC apoptosis, and eurysms (TAA) expansion rather than AAA formation, chal-
plays a critical role in multiple vascular diseases [108, 109]. lenging the role of IL-6 in AAA pathogenesis [119]. IL-17,
Expression of PKC- was dramatically enhanced in AAA generated by CD4 + T cells, facilitated AAA inflammation
tissues from human and mice [110, 111]. PKC- knockout and was crucial in AAA formation [98]. An in vitro study
mice slowed down AAA dilatation rate with a lower MCP-1 revealed that aortic SMC treated with IL-17 increasingly
expression in aortic SMC [110]. Perivascular addition of secreted proinflammatory factors such as IL-6, MCP-1 and
MCP-1 reversed this effect, suggesting that MCP-1 is a criti- TNF- [98]. Digoxin and mesenchymal stem cells are two
cal participator involved in PKC delta pathway. A mechanis- therapeutic approaches reported to modify the IL-17 pathway
tic study elucidated that PKC- activated NF-B by cytosolic [97, 120]. By far, whether anti-interleukin therapy can be
interacting with subunit p65, and thereby up-regulated MCP- brought into the clinical scenario is plausible but further in-
1 expression in AAA [112]. Unfortunately, PKC- specific vestigations are required.
inhibitor is unavailable. Further studies are needed to trans- TNF- was dramatically elevated in both human serum
late this novel finding into clinical practice. and AAA tissues, and Xiong’s study validated the role of
Receptor-Interacting Protein Kinase 3 (RIP3) is clarified TNF- in a rodent model [121]. Deficiency of TNF- inhib-
as crucial mediators of necroptosis, which is a form of well- ited CaCl2-induced AAA, preventing inflammatory cell infil-
regulated necrosis identified in past decades [113]. Recent tration and MMPs production. Intriguingly, infliximab, a
progression indicated that RIP3 is involved in aortic SMC TNF- antagonist, was also able to limit AAA expansion,
death and contributes to AAA development [114]. In human suggesting infliximab was a candidate for future AAA ther-
AAA tissues and murine elastase-induced model, expression apy [121].
of RIP3 was significantly elevated at both mRNA and pro-
tein levels [114]. Deprivation of RIP3 was resistant to AAA 3.6. Anti-Oxidative Agents
dilation, and aortic transplantation suggested that RIP3 in the Excess production of reactive oxygen species (ROS) con-
aortic wall was responsible for this protective effect. Further tinuously damages tissues and plays a vital role in many
in vitro study elucidated that knockout of RIP3 inhibited chronic diseases including AAA development [122]. Upregu-
TNF--induced SMC necroptosis, and upregulation of RIP3 lation of ROS leads to aggravation of SMC apoptosis and ele-
rendered spontaneous necroptosis. Additionally, silencing vation of MMPs activities. In AAA patients, it has already
RIP3 reduced the production of several inflammatory cyto- been reported that systemic or local oxidative stress drastically
kines including chemokine (C-C motif) ligand (CCL)-2, IL- increased [122, 123]. Accumulated evidence indicated that
6, TNF- and VCAM-1, and this pattern was partially de- oxidative stress was a therapeutic target for AAA treatment. In
pendent on amelioration of p65 phosphorylation and NF-B murine models, elimination of ROS by various anti-oxidative
activation [114]. This is the first investigation that estab- agents including vitamins, edaravone and a dipeptidyl pepti-
lished relationship between necroptosis and AAA, and it is dase (DDP)-4 inhibitor alogliptin limited AAA dilation [124-
anticipated that other molecules such as RIP1 may also be 126]. However, whether these studies can be translated into
implicated in AAA pathogenesis. Inhibition of RIP3 is an- clinical use is still elusive. Supplementation with 50 mg/day
other direction for AAA treatment. vitamin E for 5.8 years failed to change AAA incidence or
AAA rupture in a controlled, randomized trial, which chal-
3.4. MicroRNA (miRNAs) lenged the effectiveness of anti-oxidative agents in AAA ther-
MiRNAs are post-transcriptional regulators of gene and apy [127]. More clinical data is needed to verify the function.
are involved in diverse physiological and pathophysiological
processes. A review comprehensively considered recent pro- 3.7. Proteases
gress of miRNAs and AAA development [115]. MiRNA-21, - It is well established that MMPs disintegrate ECM con-
24 and -29b have been validated in experimental animal mod- tents and are involved AAA pathogenesis [128]. Deficiency
120 Current Vascular Pharmacology, 2018, Vol. 16, No. 2 Wang et al.

of MMP-2 or MMP-9, secreted by SMC or macrophage re- SMC proliferation by evoking G1 cell cycle arrest, which
spectively, significantly retarded AAA in a CaCl2 induced- was mediated by AKT phosphorylation [141]. Unfortunately,
model [129]. Development of MMPs inhibitors, such as until now, the effect of UCN2 on AAA pathogenesis has not
doxycycline abovementioned, is a temptation for future been evaluated directly in UCN2 deficiency mice. Further in
AAA therapy. However, most clinical trials of MMPs inhibi- vivo studies will supply more information about the role of
tors failed, possibly due to poor selectivity [130]. Novel UCN2 in AAA formation.
MMP-2 or MMP-9 inhibitors are promising.
It has been observed that glucose metabolism was en-
Cathepsins are types of lysosomal proteases, and when hanced in AAA patients measured by 18F-FDG uptake,
activated in acidic environment, they are speculated to poten- which is correlated with the up-regulation of glucose trans-
tiate proteolysis and contribute to AAA formation [131, porters [142]. To verify the role of glycolysis in AAA dila-
132]. Accumulated evidence demonstrated that cathepsin C, tion, Tsuruda and co-workers showed that glycolysis inhibi-
K, L, S and G were critical in AAA pathogenesis [133-137]. tor, 2-deoxyglucose, significantly attenuated the AAA for-
Lack of cathepsin C diminished AAA expansion, and neu- mation in CaCl2- and Ang II-induced mice models. Subse-
trophil infiltration and CXC-chemokine ligand (CXCL) 2 quent investigations of mechanisms revealed that 2-
were reduced [133]. Intravenous injection of wild type neu- deoxyglucose decreased expression of MCP-1, IL-1, and
trophils reversed CXCL2 generation and AAA formation in IL-6 in monocytes as well as the adherent ability of mono-
cathepsin C deficiency mice, suggesting that the influence of cytes to vascular endothelial cells [142]. Their study pro-
cathepsin C on AAA was dependent on neutrophil recruit- vided a potential novel therapeutic target. However, this
ment [133]. In elastase-induced model, absence of cathepsin needs to be further validated in the future.
L inhibited AAA development, and in parallel decreased
SMC phenotypic switching, featured by repressing SMC
MCP-1, macrophage and T-cell contents, angiogenesis and
markers and elevation of MMPs, occurs in early AAA for-
MMPs, but seldom affected SMC apoptosis [136]. The roles mation [143]. KLF4 was speculated to modulate SMC phe-
of cathepsin K in distinct murine models are still disputable
notype, and its function in AAA was affirmed by Salmon’s
[134, 138]. In an Ang II-induced AAA model, apoE-/-
group [144]. KLF4 was greatly upregulated in experimental
cathepsin K-/- knockout did not protect from AAA severity;
and human AAA, and the expression was co-localised with
compensatory activation of other proteases may account for
SMCs [144]. They created SMC-specific KLF4 knockout
this negative result [138]. Subsequently, Sun contradicted
mice and deciphered that KLF4 deficiency ameliorated AAA
that cathepsin K deficiency decreased AAA formation, ac- expansion by interacting with SMCs. Therefore, KLF4 may
companied by reduction of CD4+ T-cell aggregation and
be a potential target for AAA management. However,
SMC apoptosis [134]. Their study explained that infusion of
whether administration of KLF4 inhibitor systemically will
Ang II changed peripheral inflammatory cell phenotypes,
interfere activities of different cell types is obscure.
which obscured the function of cathepsin K in AAA. Simi-
larly, cathepsin G interfered CaCl2-induced AAA develop- TSP-1 is a pleiotropic ECM protein, which regulates a
ment, but hardly affected an Ang II-induced AAA murine body of biological activities including cell migration and
model [137]. These investigations suggested complicated angiogenesis. Recent studies correlated TSP-1 with AAA
mechanisms involved in the cathepsin family in AAA expan- pathogenesis, but the conclusion seems conflicting [145,
sion. Deprivation of cathepsin S significantly inhibited AAA 146]. TSP-1 was upregulated, evidently in adventitia, in hu-
formation and reduced SMC apoptosis and monocyte and T- man AAA specimens and experimental murine models
cell infiltration [135]. Promisingly, recent discovery of [145]. Knockout of TSP-1 limited AAA dilation in both elas-
cathepsin S inhibitor, LY3000328, was found to greatly re- tase and CaPO4-induced models, accompanied by the reduc-
duce CaCl2-induced AAA [139]. Potent, highly selective tion of macrophage aggregation and inflammatory cytokines.
inhibitors of cathepsin family represent a tempting future for Further mechanistic experiments elaborated that deficiency
AAA treatment. of TSP-1 perturbed adhesive and migratory abilities of
monocytes, which accounted for the protective effects [145].
3.8. Others Nevertheless, Krishna et al. reported that injection of LSKL,
a peptide antagonist of TSP-1, deteriorated the already-
Urocortin (UCN) is a member of corticotropin-releasing formed AAA in an Ang II-induced model [146]. In the
hormone family and also a crucial regulator of cardiovascu- meantime, administration of LSKL hampered the TGF-1
lar systems [12]. Rush’s genome expression analysis of tis- pathway, characterized by low TGF-1 and phosphorylated
sues from Ang II-induced AAA model suggested that UCN3 smad2/3 levels and expressions of TGF- receptors [146].
expression was diminished in suprarenal part of aorta, which Their work provided novel mechanism TSP-1 involved in
is vulnerable to AAA formation [140]. This study for the AAA formation, and practical delivery of TSP-1 modulators.
first time established the relationship between UCN and The role of TSP-1 in AAA is still elusive, and whether TSP-
AAA. Subsequently, their group detected UCN and its recep- 1 can be regarded as a treatment target needs to be evaluated.
tors, and found that expression of UCN2 and corticotrophin
releasing factor receptor 2(CRFR2) was significantly higher CONCLUSION
in the AAA body compared with AAA neck [141]. Immuno-
histochemical staining revealed that UCN2 expression over- We reviewed current medical treatments for AAA, and
lapped with T lymphocyte and neutrophil rich areas, suggest- there are no favourable and definitive conclusion supporting
ing that these inflammatory cells were a putative source of one or several classes of pharmacological agents. Most of the
UCN2 [141]. Likewise, plasma UCN2 levels in AAA pa- existing evidence is from retrospective and observational
tients were also elevated. In vitro, UCN2 was able to inhibit studies, and the outcomes are conflicting. Several large, ran-
Pharmacological Therapy of Abdominal Aortic Aneurysm Current Vascular Pharmacology, 2018, Vol. 16, No. 2 121

domized, placebo-controlled, double-blind trials are in pro- [12] Fraga-Silva RA, Trachet B, Stergiopulos N. Emerging pharmacol-
gress, and it is anticipated that future recommendations de- ogical treatments to prevent abdominal aortic aneurysm growth and
rupture. Curr Pharm Des 2015; 21(28): 4000-6.
rived from these trials can better guide clinical practice. [13] Van Kuijk JP, Flu WJ, Witteveen OP, Voute M, Bax JJ, Polder-
Though mechanisms of AAA formation are not fully elabo- mans D. The influence of statins on the expansion rate and rupture
rated, previous studies unravelled several dominant media- risk of abdominal aortic aneurysms. J Cardiovasc Surg (Torino)
tors and signaling pathways in AAA development, and these 2009; 50(5): 599-609.
[14] van der Meij E, Koning GG, Vriens PW, et al. A clinical evaluation
may become molecular targets for AAA control. Fascinat- of statin pleiotropy: Statins selectively and dose-dependently re-
ingly, interference of some pathways such as c-JNK even duce vascular inflammation. PLoS ONE 2013; 8(1): e53882.
leads to remission of AAA. Their agonists or antagonists are [15] Yoshimura K, Nagasawa A, Kudo J, et al. Inhibitory effect of
promising candidates for a pharmacological approach, but statins on inflammation-related pathways in human abdominal aor-
the safety, dosage, delivery and side effects should be vali- tic aneurysm tissue. Int J Mol Sci 2015; 16(5): 11213-28.
[16] Piechota-Polanczyk A, Demyanets S, Mittlboeck M, et al. The
dated in future studies. The advancements of pharmacologi- influence of simvastatin on NGAL, matrix metalloproteinases and
cal therapy may switch a surgical disease into a medical dis- their tissue inhibitors in human intraluminal thrombus and abdomi-
ease. nal aortic aneurysm tissue. Eur J Vasc Endovasc Surg 2015; 49(5):
549-55.
[17] Dunne JA, Bailey MA, Griffin KJ, Sohrabi S, Coughlin PA, Scott
CONSENT FOR PUBLICATION DJ. Statins: The holy grail of abdominal aortic aneurysm (AAA)
Not applicable. growth attenuation? A systematic review of the literature. Curr
Vasc Pharmacol 2014; 12(1): 168-72.
[18] Karrowni W, Dughman S, Hajj GP, Miller FJ. Statin therapy re-
CONFLICT OF INTEREST duces growth of abdominal aortic aneurysms. J Investig Med 2011;
59(8): 1239-43.
The authors declare no conflict of interest, financial or [19] Schouten O, van Laanen JH, Boersma E, et al. Statins are associ-
otherwise. ated with a reduced infrarenal abdominal aortic aneurysm growth.
Eur J Vasc Endovasc Surg 2006; 32(1): 21-6.
ACKNOWLEDGEMENTS [20] Periard D, Guessous I, Mazzolai L, Haesler E, Monney P, Hayoz
D. Reduction of small infrarenal abdominal aortic aneurysm expan-
This work was supported by the National Natural Science sion rate by statins. Vasa 2012; 41(1): 35-42.
[21] Takagi H, Yamamoto H, Iwata K, Goto S, Umemoto T. Effects of
Foundation of China [81470384, 81270179]. statin therapy on abdominal aortic aneurysm growth: A meta-
analysis and meta-regression of observational comparative studies.
REFERENCES Eur J Vasc Endovasc Surg 2012; 44(3): 287-92.
[22] Sweeting MJ, Thompson SG, Brown LC, Powell JT. Meta-analysis
[1] Nordon IM, Hinchliffe RJ, Loftus IM, Thompson MM. Patho- of individual patient data to examine factors affecting growth and
physiology and epidemiology of abdominal aortic aneurysms. Nat rupture of small abdominal aortic aneurysms. Br J Surg 2012;
Rev Cardiol 2011; 8(2): 92-102. 99(5): 655-65.
[2] Baxter BT, Terrin MC, Dalman RL. Medical management of small [23] Twine CP, Williams IM. Systematic review and meta-analysis of
abdominal aortic aneurysms. Circulation 2008; 117(14): 1883-9. the effects of statin therapy on abdominal aortic aneurysms. Br J
[3] Vammen S, Lindholt JS, Ostergaard L, Fasting H, Henneberg EW. Surg 2011; 98(3): 346-53.
Randomized double-blind controlled trial of roxithromycin for pre- [24] Takagi H, Matsui M, Umemoto T. A meta-analysis of clinical
vention of abdominal aortic aneurysm expansion. Br J Surg 2001; studies of statins for prevention of abdominal aortic aneurysm ex-
88(8): 1066-72. pansion. J Vasc Surg 2010; 52(6): 1675-81.
[4] Hogh A, Vammen S, Ostergaard L, Joensen JB, Henneberg EW, [25] Bergqvist D. Pharmacological interventions to attenuate the expan-
Lindholt JS. Intermittent roxithromycin for preventing progression sion of abdominal aortic aneurysm (AAA)-a systematic review. Eur
of small abdominal aortic aneurysms: Long-term results of a small J Vasc Endovasc Surg 2011; 41(5): 663-7.
clinical trial. Vasc Endovascular Surg 2009; 43(5): 452-6. [26] Stalenhoef AF. The benefit of statins in non-cardiac vascular sur-
[5] Karlsson L, Gnarpe J, Bergqvist D, Lindback J, Parsson H. The gery patients. J Vasc Surg 2009; 49(1): 260-5.
effect of azithromycin and Chlamydophilia pneumonia infection on [27] Paraskevas KI, Liapis CD, Hamilton G, Mikhailidis DP. Can stat-
expansion of small abdominal aortic aneurysms--a prospective ran- ins reduce perioperative morbidity and mortality in patients under-
domized double-blind trial. J Vasc Surg 2009; 50(1): 23-9. going non-cardiac vascular surgery? Eur J Vasc Endovasc Surg
[6] Kroon AM, Taanman JW. Clonal expansion of T cells in abdomi- 2006; 32(3): 286-93.
nal aortic aneurysm: A role for doxycycline as drug of choice? Int J [28] Paraskevas KI, Liapis CD, Hamilton G, Mikhailidis DP. Are statins
Mol Sci 2015; 16(5): 11178-95. an option in the management of abdominal aortic aneurysms? Vasc
[7] Xie X, Lu H, Moorleghen JJ, et al. Doxycycline does not influence Endovascular Surg 2008; 42(2): 128-34.
established abdominal aortic aneurysms in angiotensin II-infused [29] Wemmelund H, Hogh A, Hundborg HH, Thomsen RW, Johnsen
mice. PLoS One 2012; 7(9): e46411. SP, Lindholt JS. Statin use and rupture of abdominal aortic aneu-
[8] Meijer CA, Stijnen T, Wasser MN, Hamming JF, van Bockel JH, rysm. Br J Surg 2014; 101(8): 966-75.
Lindeman JH. Doxycycline for stabilization of abdominal aortic [30] Zhang W, Liu Z, Liu C. Effect of lipid-modifying therapy on long-
aneurysms: A randomized trial. Ann Intern Med 2013; 159(12): term mortality after abdominal aortic aneurysm repair: A systemic
815-23. review and meta-analysis. World J Surg 2015; 39(3): 794-801.
[9] Baxter BT, Matsumura J, Curci J, et al. Non-invasive treatment of [31] Galinanes EL, Reynolds S, Dombrovskiy VY, Vogel TR. The
abdominal aortic aneurysm clinical trial (N-TA(3)CT): Design of a impact of preoperative statin therapy on open and endovascular ab-
phase IIb, placebo-controlled, double-blind, randomized clinical dominal aortic aneurysm repair outcomes. Vascular 2015; 23(4):
trial of doxycycline for the reduction of growth of small abdominal 344-9.
aortic aneurysm. Contemp Clin Trials 2016; 48: 91-8. [32] de Bruin JL, Baas AF, Heymans MW, et al. Statin therapy is asso-
[10] Steinmetz EF, Buckley C, Shames ML, et al. Treatment with sim- ciated with improved survival after endovascular and open aneu-
vastatin suppresses the development of experimental abdominal rysm repair. J Vasc Surg 2014; 59(1): 39-44.
aortic aneurysms in normal and hypercholesterolemic mice. Ann [33] Kim W, Gandhi RT, Pena CS, et al. Influence of statin therapy on
Surg 2005; 241(1): 92-101. aneurysm sac regression after endovascular aortic repair. J Vasc In-
[11] Iida Y, Xu B, Schultz GM, et al. Efficacy and mechanism of angio- terv Radiol 2016; 28(1): 35-43.
tensin II receptor blocker treatment in experimental abdominal aor- [34] Raux M, Cochennec F, Becquemin JP. Statin therapy is associated
tic aneurysms. PLoS One 2012; 7(12): e49642. with aneurysm sac regression after endovascular aortic repair. J
Vasc Surg 2012; 55(6): 1587-92.
122 Current Vascular Pharmacology, 2018, Vol. 16, No. 2 Wang et al.

[35] Malekzadeh S, Fraga-Silva RA, Trachet B, Montecucco F, Mach F, pulmonary rehabilitation; national heart, lung, and blood institute;
Stergiopulos N. Role of the renin-angiotensin system on abdominal society for vascular nursing; TransAtlantic inter-society consensus;
aortic aneurysms. Eur J Clin Invest 2013; 43(12): 1328-38. and vascular disease foundation. Circulation 2006; 113(11): 463-
[36] Hackam DG, Thiruchelvam D, Redelmeier DA. Angiotensin- 654.
converting enzyme inhibitors and aortic rupture: A population- [54] Moll FL, Powell JT, Fraedrich G, et al. Management of abdominal
based case-control study. Lancet 2006; 368(9536): 659-65. aortic aneurysms clinical practice guidelines of the European soci-
[37] Thompson A, Cooper JA, Fabricius M, Humphries SE, Ashton HA, ety for vascular surgery. Eur J Vasc Endovasc Surg 2011; 41(Suppl
Hafez H. An analysis of drug modulation of abdominal aortic aneu- 1): 1-58.
rysm growth through 25 years of surveillance. J Vasc Surg 2010; [55] Lindholt JS, Sorensen HT, Michel JB, Thomsen HF, Henneberg
52(1): 55-61. EW. Low-dose aspirin may prevent growth and later surgical repair
[38] Silverberg D, Younis A, Savion N, et al. Long-term renin- of medium-sized abdominal aortic aneurysms. Vasc Endovasc Surg
angiotensin blocking therapy in hypertensive patients with normal 2008; 42(4): 329-34.
aorta may attenuate the formation of abdominal aortic aneurysms. J [56] Chen CY, Huang JW, Tzu-Chi LC, Lai WT, Huang YB. Long-term
Am Soc Hypertens 2014; 8(8): 571-7. outcome of patients with aortic aneurysms taking low-dose aspirin:
[39] Kristensen KE, Torp-Pedersen C, Gislason GH, Egfjord M, Ras- A population-based cohort study. J Investig Med 2013; 61(6):
mussen HB, Hansen PR. Angiotensin-converting enzyme inhibitors 1004-12.
and angiotensin II receptor blockers in patients with abdominal aor- [57] Wemmelund H, Jorgensen TM, Hogh A, Behr-Rasmussen C, John-
tic aneurysms: Nation-wide cohort study. Arterioscler Thromb sen SP, Lindholt JS. Low-dose aspirin and rupture of abdominal
Vasc Biol 2015; 35(3): 733-40. aortic aneurysm. J Vasc Surg 2016; 65(3): 616-25.
[40] Wemmelund H, Hogh A, Hundborg HH, Johnsen SP, Lindholt JS. [58] The efficacy of Ticagrelor on abdominal aortic aneurysm (AAA)
Preadmission use of renin-angiotensin blockers and rupture of ab- expansion. [Online] 2014 [cited 2017]. Available at: (https:
dominal aortic aneurysm: A nationwide, population-based study. //clinicaltrials.gov/ct2/show/NCT02070653).
Pharmacoepidemiol Drug Saf 2016; 25(2): 141-50. [59] Shi GP, Lindholt JS. Mast cells in abdominal aortic aneurysms.
[41] Sweeting MJ, Thompson SG, Brown LC, Greenhalgh RM, Powell Curr Vasc Pharmacol 2013; 11(3): 314-26.
JT. Use of angiotensin converting enzyme inhibitors is associated [60] Sun J, Sukhova GK, Yang M, et al. Mast cells modulate the patho-
with increased growth rate of abdominal aortic aneurysms. J Vasc genesis of elastase-induced abdominal aortic aneurysms in mice. J
Surg 2010; 52(1): 1-4. Clin Invest 2007; 117(11): 3359-68.
[42] Railton CJ, Wolpin J, Lam-McCulloch J, Belo SE. Renin- [61] Tsuruda T, Kato J, Hatakeyama K, et al. Adventitial mast cells
angiotensin blockade is associated with increased mortality after contribute to pathogenesis in the progression of abdominal aortic
vascular surgery. Can J Anaesth 2010; 57(8): 736-44. aneurysm. Circ Res 2008; 102(11): 1368-77.
[43] Bicknell CD, Kiru G, Falaschetti E, Powell JT, Poulter NR. An [62] Wang Y, Shi GP. Mast cell chymase and tryptase in abdominal
evaluation of the effect of an angiotensin-converting enzyme in- aortic aneurysm formation. Trends Cardiovasc Med 2012; 22(6):
hibitor on the growth rate of small abdominal aortic aneurysms: A 150-5.
randomized placebo-controlled trial (AARDVARK). Eur Heart J [63] Sillesen H, Eldrup N, Hultgren R, et al. Randomized clinical trial
2016; 37(42): 3213-21. of mast cell inhibition in patients with a medium-sized abdominal
[44] Ronald LD. Study of the effectiveness of Telmisartan in slowing aortic aneurysm. Br J Surg 2015; 102(8): 894-901.
the progression of abdominal aortic aneurysms (TEDY). [Online] [64] Nakashima H, Aoki M, Miyake T, et al. Inhibition of experimental
2012 [cited 2017]. Available at: (https: abdominal aortic aneurysm in the rat by use of decoy oligode-
//clinicaltrials.gov/show/NCT01683084). oxynucleotides suppressing activity of nuclear factor kappaB and
[45] Ketha SS, Warrington KJ, McPhail IR. Inflammatory abdominal ets transcription factors. Circulation 2004; 109(1): 132-8.
aortic aneurysm: A case report and review of literature. Vasc En- [65] Miyake T, Aoki M, Masaki H, et al. Regression of abdominal
dovascular Surg 2014; 48(1): 65-9. aortic aneurysms by simultaneous inhibition of nuclear factor kap-
[46] Yabe T, Hamada T, Kubo T, et al. Inflammatory abdominal aortic paB and ets in a rabbit model. Circ Res 2007; 101(11): 1175-84.
aneurysm successfully treated with steroid therapy. J Am Coll Car- [66] Miyake T, Aoki M, Nakashima H, et al. Prevention of abdominal
diol 2010; 55(25): 2877. aortic aneurysms by simultaneous inhibition of NFkappaB and ets
[47] Fatima J, Gota C, Clair DG, Billings SD, Gornik HL. Inflammatory using chimeric decoy oligonucleotides in a rabbit model. Gene
abdominal aortic aneurysm with retroperitoneal fibrosis. Circula- Ther 2006; 13(8): 695-704.
tion 2014; 130(15): 1300-2. [67] Tai H, Tsai P, Chen J, et al. Peroxisome proliferator-activated
[48] de la Motte L, Kehlet H, Vogt K, et al. Preoperative methylpredni- receptor  level contributes to structural integrity and component
solone enhances recovery after endovascular aortic repair: A ran- production of elastic fibers in the aorta. Hypertension 2016; 67(6):
domized, double-blind, placebo-controlled clinical trial. Ann Surg 1298-308.
2014; 260(3): 540-9. [68] Jones A, Deb R, Torsney E, et al. Rosiglitazone reduces the devel-
[49] Lindholt JS, Bjorck M, Michel JB. Anti-platelet treatment of mid- opment and rupture of experimental aortic aneurysms. Circulation
dle-sized abdominal aortic aneurysms. Curr Vasc Pharmacol 2013; 2009; 119(24): 3125-32.
11(3): 305-13. [69] Golledge J, Cullen B, Rush C, et al. Peroxisome proliferator-
[50] Parr A, McCann M, Bradshaw B, Shahzad A, Buttner P, Golledge activated receptor ligands reduce aortic dilatation in a mouse model
J. Thrombus volume is associated with cardiovascular events and of aortic aneurysm. Atherosclerosis 2010; 210(1): 51-6.
aneurysm growth in patients who have abdominal aortic aneu- [70] Motoki T, Kurobe H, Hirata Y, et al. PPAR-gamma agonist attenu-
rysms. J Vasc Surg 2011; 53(1): 28-35. ates inflammation in aortic aneurysm patients. Gen Thorac Cardio-
[51] Owens AR, Edwards TL, Antoniak S, et al. Platelet inhibitors vasc Surg 2015; 63(10): 565-71.
reduce rupture in a mouse model of established abdominal aortic [71] Hamblin M, Chang L, Zhang H, Yang K, Zhang J, Chen YE. Vas-
aneurysm. Arterioscler Thromb Vasc Biol 2015; 35(9): 2032-41. cular smooth muscle cell peroxisome proliferator-activated recep-
[52] Liu O, Jia L, Liu X, et al. Clopidogrel, a platelet P2Y12 receptor tor-gamma deletion promotes abdominal aortic aneurysms. J Vasc
inhibitor, reduces vascular inflammation and angiotensin II in- Surg 2010; 52(4): 984-93.
duced-abdominal aortic aneurysm progression. PLoS ONE 2012; [72] Nakashiro S, Matoba T, Umezu R, et al. Pioglitazone-incorporated
7(12): e51707. nanoparticles prevent plaque destabilization and rupture by regulat-
[53] Hirsch AT, Haskal ZJ, Hertzer NR, et al. ACC/AHA 2005 practice ing monocyte/macrophage differentiation in ApoE-/- Mice. Arterio-
guidelines for the management of patients with peripheral arterial scler Thromb Vasc Biol 2016; 36(3): 491-500.
disease (lower extremity, renal, mesenteric, and abdominal aortic): [73] Ghosh A, Lu G, Su G, et al. Phosphorylation of AKT and abdomi-
A collaborative report from the American association for vascular nal aortic aneurysm formation. Am J Pathol 2014; 184(1): 148-58.
surgery/society for vascular surgery, society for cardiovascular an- [74] Shen YH, Zhang L, Ren P, et al. AKT2 confers protection against
giography and interventions, society for vascular medicine and bi- aortic aneurysms and dissections. Circ Res 2013; 112(4): 618-32.
ology, society of interventional radiology, and the ACC/AHA task [75] Krishna SM, Seto SW, Moxon JV, et al. Fenofibrate increases
force on practice guidelines (writing committee to develop guide- high-density lipoprotein and sphingosine 1 phosphate concentra-
lines for the management of patients with peripheral arterial Dis- tions limiting abdominal aortic aneurysm progression in a mouse
ease): Endorsed by the American association of cardiovascular and model. Am J Pathol 2012; 181(2): 706-18.
Pharmacological Therapy of Abdominal Aortic Aneurysm Current Vascular Pharmacology, 2018, Vol. 16, No. 2 123

[76] Gomez I, Foudi N, Longrois D, Norel X. The role of prostaglandin production and inhibit abdominal aortic aneurysms. Arterioscler
E2 in human vascular inflammation. Prostaglandins Leukot Essent Thromb Vasc Biol 2016; 36(5): 908-18.
Fatty Acids 2013; 89(2-3): 55-63. [98] Sharma AK, Lu G, Jester A, et al. Experimental abdominal aortic
[77] Bayston T, Ramessur S, Reise J, Jones KG, Powell JT. Prostaglan- aneurysm formation is mediated by IL-17 and attenuated by mes-
din E2 receptors in abdominal aortic aneurysm and human aortic enchymal stem cell treatment. Circulation 2012; 126(11_suppl_1):
smooth muscle cells. J Vasc Surg 2003; 38(2): 354-9. S38-45.
[78] Yokoyama U, Ishiwata R, Jin MH, et al. Inhibition of EP4 signal- [99] Meng X, Yang J, Zhang K, et al. Regulatory T cells prevent angio-
ing attenuates aortic aneurysm formation. PLoS One 2012; 7(5): tensin II-induced abdominal aortic aneurysm in apolipoprotein E
e36724. knockout mice. Hypertension 2014; 64(4): 875-82.
[79] Cao RY, St AT, Li X, et al. Prostaglandin receptor EP4 in abdomi- [100] Zhou Y, Wu W, Lindholt JS, et al. Regulatory T cells in human
nal aortic aneurysms. Am J Pathol 2012; 181(1): 313-21. and angiotensin II-induced mouse abdominal aortic aneurysms.
[80] Camacho M, Dilme J, Sola-Villa D, et al. Microvascular COX- Cardiovasc Res 2015; 107(1): 98-107.
2/mPGES-1/EP-4 axis in human abdominal aortic aneurysm. J [101] Dale MA, Xiong W, Carson JS, et al. Elastin-derived peptides
Lipid Res 2013; 54(12): 3506-15. promote abdominal aortic aneurysm formation by modulating
[81] Tang EH, Shvartz E, Shimizu K, et al. Deletion of EP4 on bone M1/M2 macrophage polarization. J Immunol 2016; 196(11): 4536-
marrow-derived cells enhances inflammation and angiotensin II- 43.
induced abdominal aortic aneurysm formation. Arterioscler [102] Yoshimura K, Aoki H, Ikeda Y, et al. Regression of abdominal
Thromb Vasc Biol 2011; 31(2): 261-9. aortic aneurysm by inhibition of c-Jun N-terminal kinase. Nat Med
[82] Hans CP, Koenig SN, Huang N, et al. Inhibition of Notch1 signal- 2005; 11(12): 1330-8.
ing reduces abdominal aortic aneurysm in mice by attenuating [103] Chen F, Zhang Z, Zhu X. Inhibition of development of experimen-
macrophage-mediated inflammation. Arterioscler Thromb Vasc tal abdominal aortic aneurysm by c-Jun N-terminal protein kinase
Biol 2012; 32(12): 3012-23. inhibitor combined with LYSYL oxidase gene modified smooth
[83] Cheng J, Koenig SN, Kuivaniemi HS, Garg V, Hans CP. Pharma- muscle progenitor cells. Eur J Pharmacol 2015; 766: 114-21.
cological inhibitor of notch signaling stabilizes the progression of [104] Guo ZZ, Cao QA, Li ZZ, et al. SP600125 attenuates nicotine-
small abdominal aortic aneurysm in a mouse model. J Am Heart related aortic aneurysm formation by inhibiting matrix metallopro-
Assoc 2014; 3(6): e1064. teinase production and CC chemokine-mediated macrophage mi-
[84] Zheng YH, Li FD, Tian C, Ren HL, Du J, Li HH. Notch gamma- gration. Mediators Inflamm 2016; 2016: 9142425.
secretase inhibitor dibenzazepine attenuates angiotensin II-induced [105] Ghosh A, DiMusto PD, Ehrlichman LK, et al. The role of extracel-
abdominal aortic aneurysm in ApoE knockout mice by multiple lular signal-related kinase during abdominal aortic aneurysm for-
mechanisms. PLoS One 2013; 8(12): e83310. mation. J Am Coll Surgeons 2012; 215(5): 668-80.
[85] Wang YW, Ren HL, Wang HF, Li FD, Li HH, Zheng YH. Com- [106] Groeneveld ME, van Burink MV, Begieneman MPV, et al. Activa-
bining detection of Notch1 and tumor necrosis factor-alpha con- tion of extracellular signal-related kinase in abdominal aortic aneu-
verting enzyme is a reliable biomarker for the diagnosis of abdomi- rysm. Eur J Clin Invest 2016; 46(5): 440-7.
nal aortic aneurysms. Life Sci 2015; 127: 39-45. [107] Cheng ZB, Shao WW, Liu YN, et al. Extracellular signal-regulated
[86] Doyle AJ, Redmond EM, Gillespie DL, et al. Differential expres- kinases (ERK) inhibitors from Aristolochia yunnanensis. J Nat
sion of Hedgehog/Notch and transforming growth factor-beta in Prod 2013; 76(4): 664-71.
human abdominal aortic aneurysms. J Vasc Surg 2015 ; 62(2): 464- [108] Si Y, Ren J, Wang P, et al. Protein kinase C-delta mediates adven-
70. titial cell migration through regulation of monocyte chemoattrac-
[87] Owens AR, Rateri DL, Howatt DA, et al. MyD88 deficiency at- tant protein-1 expression in a rat angioplasty model. Arterioscler
tenuates angiotensin II-induced abdominal aortic aneurysm forma- Thromb Vasc Biol 2012; 32(4): 943-54.
tion independent of signaling through Toll-like receptors 2 and 4. [109] Leitges M, Mayr M, Braun U, et al. Exacerbated vein graft arterio-
Arterioscler Thromb Vasc Biol 2011; 31(12): 2813-9. sclerosis in protein kinase C delta-null mice. J Clin Invest 2001;
[88] Lai CH, Wang KC, Lee FT, et al. Toll-like receptor 4 is essential in 108(10): 1505-12.
the development of abdominal aortic aneurysm. PLoS One 2016; [110] Morgan S, Yamanouchi D, Harberg C, et al. Elevated protein
11(1): e146565. kinase C-delta contributes to aneurysm pathogenesis through
[89] Qin Z, Bagley J, Sukhova G, et al. Angiotensin II-induced TLR4 stimulation of apoptosis and inflammatory signaling. Arterioscler
mediated abdominal aortic aneurysm in apolipoprotein E knockout Thromb Vasc Biol 2012; 32(10): 2493-502.
mice is dependent on STAT3. J Mol Cell Cardiol 2015; 87: 160-70. [111] Schubl S, Tsai S, Ryer EJ, et al. Upregulation of protein kinase
[90] Huggins C, Pearce S, Peri F, Neumann F, Cockerill G, Pirianov G. cdelta in vascular smooth muscle cells promotes inflammation in
A novel small molecule TLR4 antagonist (IAXO-102) negatively abdominal aortic aneurysm. J Surg Res 2009; 153(2): 181-7.
regulates non-hematopoietic toll like receptor 4 signalling and in- [112] Ren J, Wang Q, Morgan S, et al. Protein kinase C-(PKC) regu-
hibits aortic aneurysms development. Atherosclerosis 2015; 242(2): lates proinflammatory chemokine expression through cytosolic in-
563-70. teraction with the NF-B subunit p65 in vascular smooth muscle
[91] Yan H, Cui B, Zhang X, et al. Antagonism of toll-like receptor 2 cells. J Biol Chem 2014; 289(13): 9013-26.
attenuates the formation and progression of abdominal aortic aneu- [113] Linkermann A, Green DR. Necroptosis. New Engl J Med 2014;
rysm. Acta Pharm Sin B 2015; 5(3): 176-87. 370(5): 455-65.
[92] Yamawaki-Ogata A. Mesenchymal stem cells for treatment of [114] Wang Q, Liu Z, Ren J, Morgan S, Assa C, Liu B. Receptor-
aortic aneurysms. World J Stem Cells 2014; 6(3): 278. interacting protein kinase 3 contributes to abdominal aortic aneu-
[93] Hashizume R, Yamawaki-Ogata A, Ueda Y, Wagner WR, Narita rysms via smooth muscle cell necrosis and inflammation. Circ Res
Y. Mesenchymal stem cells attenuate angiotensin II-induced aortic 2015; 116(4): 600-11.
aneurysm growth in apolipoprotein E-deficient mice. J Vasc Surg [115] Raffort J, Lareyre F, Clement M, Mallat Z. Micro-RNAs in ab-
2011; 54(6): 1743-52. dominal aortic aneurysms: Insights from animal models and rele-
[94] Fu XM, Yamawaki-Ogata A, Oshima H, Ueda Y, Usui A, Narita vance to human disease. Cardiovasc Res 2016; 110(2): 165-77.
Y. Intravenous administration of mesenchymal stem cells prevents [116] Johnston WF, Salmon M, Su G, et al. Genetic and pharmacologic
angiotensin II-induced aortic aneurysm formation in apolipoprotein disruption of interleukin-1beta signaling inhibits experimental aor-
E-deficient mouse. J Transl Med 2013; 11: 175. tic aneurysm formation. Arterioscler Thromb Vasc Biol 2013;
[95] Schneider F, Saucy F, de Blic R, et al. Bone marrow mesenchymal 33(2): 294-304.
stem cells stabilize already-formed aortic aneurysms more effi- [117] Dawson J, Cockerill GW, Choke E, Belli A, Loftus I, Thompson
ciently than vascular smooth muscle cells in a rat model. Eur J MM. Aortic aneurysms secrete interleukin-6 into the circulation. J
Vasc Endovasc Surg 2013; 45(6): 666-72. Vasc Surg 2007; 45(2): 350-6.
[96] Riera DML, Largo C, Ramirez JR, et al. Potential of mesenchymal [118] Harrison SC, Smith AJ, Jones GT, et al. Interleukin-6 receptor
stem cell in stabilization of abdominal aortic aneurysm sac. J Surg pathways in abdominal aortic aneurysm. Eur Heart J 2013; 34(48):
Res 2015; 195(1): 325-33. 3707-16.
[97] Sharma AK, Salmon MD, Lu G, et al. Mesenchymal stem cells [119] Pope NH, Salmon M, Johnston WF, et al. Interleukin-6 receptor
attenuate NADPH oxidase-dependent high mobility group box 1 inhibition prevents descending thoracic aortic aneurysm formation.
Ann Thorac Surg 2015; 100(5): 1620-6.
124 Current Vascular Pharmacology, 2018, Vol. 16, No. 2 Wang et al.

[120] Wei Z, Wang Y, Zhang K, et al. Inhibiting the Th17/IL-17A- [135] Qin Y, Cao X, Guo J, et al. Deficiency of cathepsin S attenuates
related inflammatory responses with digoxin confers protection angiotensin II-induced abdominal aortic aneurysm formation in
against experimental abdominal aortic aneurysm. Arterioscler apolipoprotein E-deficient mice. Cardiovasc Res 2012; 96(3): 401-
Thromb Vasc Biol 2014; 34(11): 2429-38. 10.
[121] Xiong W, MacTaggart J, Knispel R, Worth J, Persidsky Y, Baxter [136] Sun J, Sukhova GK, Zhang J, et al. Cathepsin L activity is essential
BT. Blocking TNF-alpha attenuates aneurysm formation in a mur- to elastase perfusion-induced abdominal aortic aneurysms in mice.
ine model. J Immunol 2009; 183(4): 2741-6. Arterioscler Thromb Vasc Biol 2011; 31(11): 2500-8.
[122] Emeto TI, Moxon JV, Au M, Golledge J. Oxidative stress and [137] Wang J, Sukhova GK, Liu J, et al. Cathepsin G deficiency reduces
abdominal aortic aneurysm: Potential treatment targets. Clin Sci periaortic calcium chloride injury-induced abdominal aortic aneu-
(Lond) 2016; 130(5): 301-15. rysms in mice. J Vasc Surg 2015; 62(6): 1615-24.
[123] Mentese U, Turan I, Usta S, et al. Systemic oxidant/antioxidant [138] Bai L, Beckers L, Wijnands E, et al. Cathepsin K gene disruption
balance in human abdominal aortic aneurysm. Perfusion 2016; does not affect murine aneurysm formation. Atherosclerosis 2010;
31(4): 288-94. 209(1): 96-103.
[124] Shang T, Liu Z, Liu CJ. Antioxidant vitamin C attenuates experi- [139] Jadhav PK, Schiffler MA, Gavardinas K, et al. Discovery of
mental abdominal aortic aneurysm development in an elastase- cathepsin S inhibitor LY3000328 for the treatment of abdominal
induced rat model. J Surg Res 2014; 188(1): 316-25. aortic aneurysm. ACS Med Chem Lett 2014; 5(10): 1138-42.
[125] Bao W, Morimoto K, Hasegawa T, et al. Orally administered [140] Rush C, Nyara M, Moxon JV, Trollope A, Cullen B, Golledge J.
dipeptidyl peptidase-4 inhibitor (alogliptin) prevents abdominal Whole genome expression analysis within the angiotensin II-
aortic aneurysm formation through an antioxidant effect in rats. J apolipoprotein E deficient mouse model of abdominal aortic aneu-
Vasc Surg 2014; 59(4): 1098-108. rysm. BMC Genomics 2009; 10: 298.
[126] Morimoto K, Hasegawa T, Tanaka A, et al. Free-radical scavenger [141] Emeto TI, Moxon JV, Biros E, et al. Urocortin 2 is associated with
edaravone inhibits both formation and development of abdominal abdominal aortic aneurysm and mediates anti-proliferative effects
aortic aneurysm in rats. J Vasc Surg 2012; 55(6): 1749-58. on vascular smooth muscle cells via corticotrophin releasing factor
[127] Tornwall ME, Virtamo J, Haukka JK, Albanes D, Huttunen JK. receptor 2. Clin Sci (Lond) 2014; 126(7): 517-27.
Alpha-tocopherol (vitamin E) and beta-carotene supplementation [142] Tsuruda T, Hatakeyama K, Nagamachi S, et al. Inhibition of devel-
does not affect the risk for large abdominal aortic aneurysm in a opment of abdominal aortic aneurysm by glycolysis restriction. Ar-
controlled trial. Atherosclerosis 2001; 157(1): 167-73. terioscler Thromb Vasc Biol 2012; 32(6): 1410-7.
[128] Maegdefessel L, Dalman RL, Tsao PS. Pathogenesis of abdominal [143] Ailawadi G, Moehle CW, Pei H, et al. Smooth muscle phenotypic
aortic aneurysms: microRNAs, proteases, genetic associations. modulation is an early event in aortic aneurysms. J Thorac Cardio-
Annu Rev Med 2014; 65: 49-62. vasc Surg 2009; 138(6): 1392-9.
[129] Longo GM, Xiong W, Greiner TC, Zhao Y, Fiotti N, Baxter BT. [144] Salmon M, Johnston WF, Woo A, et al. KLF4 regulates abdominal
Matrix metalloproteinases 2 and 9 work in concert to produce aor- aortic aneurysm morphology and deletion attenuates aneurysm
tic aneurysms. J Clin Invest 2002; 110(5): 625-32. formation. Circulation 2013; 128(Suppl 1): 163-74.
[130] Dorman G, Cseh S, Hajdu I, et al. Matrix metalloproteinase inhibi- [145] Liu Z, Morgan S, Ren J, et al. Thrombospondin-1 (TSP1) contrib-
tors: a critical appraisal of design principles and proposed therapeu- utes to the development of vascular inflammation by regulating
tic utility. Drugs 2010; 70(8): 949-64. monocytic cell motility in mouse models of abdominal aortic aneu-
[131] Wang Y, Tang C, Qin Y. Cathepsins: A new culprit behind ab- rysm. Circ Res 2015; 117(2): 129-41.
dominal aortic aneurysm. Regen Med Res 2013; 1(1): 5. [146] Krishna SM, Seto SW, Jose RJ, et al. A peptide antagonist of
[132] Qin Y, Cao X, Yang Y, Shi GP. Cysteine protease cathepsins and thrombospondin-1 promotes abdominal aortic aneurysm progres-
matrix metalloproteinases in the development of abdominal aortic sion in the angiotensin II-infused apolipoprotein-E-deficient
aneurysms. Future Cardiol 2013; 9(1): 89-103. mouse. Arterioscler Thromb Vasc Biol 2015; 35(2): 389-98.
[133] Pagano MB, Bartoli MA, Ennis TL, et al. Critical role of dipeptidyl [147] Wilmink ABM, Vardulaki KA, Hubbard CSF, et al. Are antihyper-
peptidase I in neutrophil recruitment during the development of ex- tensive drugs associated with abdominal aortic aneurysms? J Vasc
perimental abdominal aortic aneurysms. Proc Natl Acad Sci USA Surg 2002; 36(4): 751-7.
2007; 104(8): 2855-60. [148] Ferguson CD, Clancy P, Bourke B, et al. Association of statin
[134] Sun J, Sukhova GK, Zhang J, et al. Cathepsin K deficiency reduces prescription with small abdominal aortic aneurysm progression.
elastase perfusion-induced abdominal aortic aneurysms in mice. Am Heart J 2010; 159(2): 307-13.
Arterioscler Thromb Vasc Biol 2012; 32(1): 15-23.

You might also like