ArticlePDF AvailableLiterature Review

Pharmacologic Management of Aneurysms

Authors:

Abstract and Figures

Current management of aortic aneurysms relies exclusively on prophylactic operative repair of larger aneurysms. Great potential exists for successful medical therapy that halts or reduces aneurysm progression and hence alleviates or postpones the need for surgical repair. Preclinical studies in the context of abdominal aortic aneurysm identified hundreds of candidate strategies for stabilization, and data from preoperative clinical intervention studies show that interventions in the pathways of the activated inflammatory and proteolytic cascades in enlarging abdominal aortic aneurysm are feasible. Similarly, the concept of pharmaceutical aorta stabilization in Marfan syndrome is supported by a wealth of promising studies in the murine models of Marfan syndrome-related aortapathy. Although some clinical studies report successful medical stabilization of growing aortic aneurysms and aortic root stabilization in Marfan syndrome, these claims are not consistently confirmed in larger and controlled studies. Consequently, no medical therapy can be recommended for the stabilization of aortic aneurysms. The discrepancy between preclinical successes and clinical trial failures implies shortcomings in the available models of aneurysm disease and perhaps incomplete understanding of the pathological processes involved in later stages of aortic aneurysm progression. Preclinical models more reflective of human pathophysiology, identification of biomarkers to predict severity of disease progression, and improved design of clinical trials may more rapidly advance the opportunities in this important field.
Content may be subject to copyright.
631
An aneurysm is a localized dilatation of larger blood ves-
sels that is related to regional weakening of the wall
structure.1 Although the large majority of aneurysms presents
within arterial tree, venous aneurysms do occur. Aneurysms
are generally associated with rupture and a life-threatening
hemorrhage, yet some aneurysms (in particular, popliteal2 and
venous aneurysms)3 typically manifest through symptoms of
acute thrombosis and embolism.
There are several classification systems for aneurysms.
From the perspective of medical therapy, the most help-
ful attribution is that of primary and secondary aneurysms.
Primary aneurysms relate to a matrix defect in vessel wall
(ie, fibrillin deficiency in Marfan syndrome, collagen III de-
ficiency in the vascular type Ehlers Danlos syndrome,4 and
unknown defect[s] in aneurysms associated with bicuspid a-
ortic valves).5,6 Secondary aneurysms relate to extensive ma-
trix turnover and pathological vessel wall remodeling5,7 in
response to a primary inflammatory insult (ie, infection, im-
mune diseases [Kawasaki syndrome, giant cell arthritis, and
Behçet syndrome],8 and the degenerative abdominal aortic an-
eurysm [AAA]).1
Although aneurysms occur throughout the vascular tree,
there is a remarkable topographical distribution for most an-
eurysms (ie, descending thoracic aorta for giant cell arteritis,
infrarenal aorta in AAA disease, etc).9 Although this may be
caused by local hemodynamic patterns and associated wall
stresses, it likely could reflect the different embryological ori-
gins of the vascular tree,9 which result in a persistent regional
diversity in microvascular endothelium,10 mesenchymal cell
characteristics,11–13 and immunologic makeup.14,15 The remark-
able regional diversity in susceptibility is clearly illustrated by
the iliac trajectory: unlike the adjacent common iliac, internal
iliac, and common femoral arteries, the external iliac artery is
remarkably resistant to degenerative aneurysms.
The focus of this compendium will be on the perspectives
for medical therapy in the context of AAA and aneurysms as-
sociated with Marfan syndrome as clinical intervention data
are available for these aneurysms. Systemic immune suppres-
sion in the context of autoimmune diseases, such as Kawasaki
syndrome and giant cell arteritis, and antibiotic strategies for
infection-related aneurysms (Q fever, bacterial) are beyond
the scope of this article.
Abdominal Aortic Aneurysm
An AAA is a localized dilatation caused by segmental weak-
ening of the terminal aorta segment. The prevalence of the
disease depends on the population studied, with reported prev-
alences varying between 1.4% and 12.4%.16 The disease car-
ries a complex genetic predisposition17,18 and predominantly
affects elderly men with a history of smoking.19
AAAs are generally asymptomatic and are usually diag-
nosed by screening or as an incidental finding. The natural
From the Department of Vascular Surgery, Leiden University Medical Center, the Netherlands (J.H.L.); and Division of Vascular Surgery, School of
Medicine and Public Health, University of Wisconsin, Madison (J.S.M.).
Correspondence to Jan H. Lindeman, MD, PhD, Department of Vascular Surgery, Leiden University Medical Center, K6-r, PO Box 9600, 2300 RC,
Leiden, the Netherlands. Email lindeman@lumc.nl
Aortic Aneurysms Compendium
© 2019 American Heart Association, Inc.
Abstract: Current management of aortic aneurysms relies exclusively on prophylactic operative repair of larger
aneurysms. Great potential exists for successful medical therapy that halts or reduces aneurysm progression and
hence alleviates or postpones the need for surgical repair. Preclinical studies in the context of abdominal aortic
aneurysm identified hundreds of candidate strategies for stabilization, and data from preoperative clinical
intervention studies show that interventions in the pathways of the activated inflammatory and proteolytic cascades
in enlarging abdominal aortic aneurysm are feasible. Similarly, the concept of pharmaceutical aorta stabilization in
Marfan syndrome is supported by a wealth of promising studies in the murine models of Marfan syndrome–related
aortapathy. Although some clinical studies report successful medical stabilization of growing aortic aneurysms and
aortic root stabilization in Marfan syndrome, these claims are not consistently confirmed in larger and controlled
studies. Consequently, no medical therapy can be recommended for the stabilization of aortic aneurysms. The
discrepancy between preclinical successes and clinical trial failures implies shortcomings in the available models
of aneurysm disease and perhaps incomplete understanding of the pathological processes involved in later stages
of aortic aneurysm progression. Preclinical models more reflective of human pathophysiology, identification of
biomarkers to predict severity of disease progression, and improved design of clinical trials may more rapidly advance
the opportunities in this important field. (Circ Res. 2019;124:631-646. DOI: 10.1161/CIRCRESAHA.118.312439.)
Key Words: aneurysm aortic aneurysm, abdominal Marfan syndrome mice models, animal
review literature as topic therapy, drug
Pharmacologic Management of Aneurysms
Jan H. Lindeman, Jon S. Matsumura
Circulation Research is available at https://www.ahajournals.org/journal/res DOI: 10.1161/CIRCRESAHA.118.312439
Downloaded from http://ahajournals.org by on February 20, 2019
632 Circulation Research February 15, 2019
history of the disease is that of slow progression and ultimate
rupture.20 Ruptured AAA is a dramatic catastrophe, and aor-
tic emergencies constitute one of the leading causes of acute
death in elderly men.1,16 Risk of rupture is minimal in small
aneurysms (ie, <50 mm) but progressively increases with en-
larging AAA size; estimated annual rupture risks are <1% for
AAA with a diameter of 50 mm to >30% for an AAA exceed-
ing 80 mm diameter.21
AAA management has been centered for decades on sur-
gical repair of larger aneurysms to mitigate the risks of rup-
ture. Multiple trials have shown no benefit of repair of AAA at
sizes <55 mm diameter, and consequently, current guidelines
advise watchful waiting for aneurysms <55 mm and preven-
tive repair once the AAA grows >55 mm,1,19,21 possibly with
a slightly lower intervention threshold for repair in women.22
The 2 surgical options for repair are open repair (through
a transperitoneal or retroperitoneal approach) or endovas-
cular aneurysm repair (EVAR; through a transarterial ap-
proach).19,21 Decisions for the type of repair are dictated
by AAA-specific features, such as neck characteristics and
proximity to major important branches, as well as by pa-
tients’ preferences and characteristics, such as frailty and o-
besity.19,21 The majority of patients is currently managed by
EVAR. Open repair comes with significant higher periopera-
tive mortality and morbidity; registry-based studies report
30-day mortality rates of 4% to 5% for men and 6% to 8%
for women,22,23 and perioperative morbidity of open repair
is considerable.19 However, open repair has an established
long-term durability, although incisional hernia remains a
common cause of late reintervention.19 EVAR has superior
short-term outcomes but comes with higher rates of aortic
reintervention and possibly higher costs.24 Moreover, there is
emerging concern in the published literature about the mid-
and long-term durability of EVAR with possibly excess late
mortality in patients who received EVAR.25,26
Considering the fact that the sole indication for elective
AAA repair is rupture prevention,19 it has been pointed out
that medical stabilization of small diameter aneurysms, keeps
small aneurysms small and thereby prevents or reduces the
need for surgical repair, could be advantageous. This has nat-
ural appeal to patients and from an economic point of view.27
Moreover, medical aneurysm stabilization could be benefi-
cial as add-on strategy in patients considered at high risk for
endoleak.28 It is conceivable that in patients who have aneu-
rysm neck prone to dilation, stabilization of the neck could
reduce the incidence of late type Ia endoleak. All in all, med-
ical AAA stabilization has been brought forward as an unmet
medical need.29
Targets for Medical AAA Therapy
Candidate targets for therapy are dictated by the prevailing
concepts of the processes driving AAA disease progression.
It is generally assumed that AAA progression is driven by a
localized inflammatory response and an accompanying pro-
teolytic imbalance.20,30 Consequently, proposed interventions
directly or indirectly aim at targeting aspects of the inflamma-
tory response or at rectifying the proteolytic imbalance. The
pertinence of these strategies is supported by a wealth of pre-
clinical studies. The vast majority of these are performed in the
standard rodent models of AAA disease: the elastase model,
the CaCl2 model, or the angiotensin/LDLR−/− (low-density li-
poprotein receptor) model.31,32
The elastase model is based on a transient exposure (gen-
erally brief intraaortic exposure) of an isolated infrarenal aorta
segment with porcine pancreatic elastase.33 The rationale be-
hind the model is the notion that loss of elastin is one of the
most notable features of AAA disease. Yet, although the di-
sease is undoubtedly characterized by extensive loss of elas-
tin, it is important to point out that loss of elastin per se is not
responsible for the critical wall failure in AAA. First of all,
loss of elastin is an early phenomenon in clinical AAA devel-
opment, and the elastolysis is virtually complete before the di-
sease reaches the critical 55-mm diameter threshold.34 Second,
clinical experience shows that chemical or surgical removal of
elastin (endarteriectomy) does not result in aneurysm forma-
tion.35,36 The validity of this clinical observation is supported
by experimental data that show that although elastin critically
contributes to the elastic recoil of the aortic wall, it does not
contribute to the resilience of the wall. In fact, studies by
Dobrin et al37 show that the resilience of the wall essentially re-
lies on vascular collagen. This phenomenon is also reflected in
the dynamics of the elastase model in which exposure to pure
elastin does not immediately induce AAA formation38 and in
which the initial response following porcine pancreatic elastin
preparations is a small increase in aortic diameter, presumably
reflecting loss of elastic recoil.39 The actual aneurysm forma-
tion is secondary and reflects a delayed, secondary response,
elicited by the elastase-induced inflammatory response.37
Although the model is referred as the elastase model, ex-
posure to pure pancreatic elastase does not elicit aneurysm
formation.38 As such, contaminants of the porcine pancre-
atic elastase preparation appear crucial for AAA induction.
Further, the model is dependent on the genetic background
of the mouse with a strict requirement for strains with Th1-
dominated inflammatory responses,39 underscoring the rele-
vance of the inflammatory response in model.
Nonstandard Abbreviations and Acronyms
AAA abdominal aortic aneurysm
ACE angiotensin-converting enzyme
ADAM Aneurysm Detection and Management
ATR1 angiotensin II receptor type 1
EVAR endovascular aneurysm repair
FBN1 fibrillin-1
IL interleukin
LDLR low-density lipoprotein receptor
LDL low-density lipoprotein
LOX lysyl oxidase
MMP matrix metalloproteinase
N-TA^3CT Noninvasive Treatment of Abdominal Aortic Aneurysm Clinical
Trial
NFAT2 nuclear factor of activated T cells-2
PPAR peroxisome proliferator–activated receptor
TGF-β transforming growth factor-β
TNF-α tumor necrosis factor-α
Downloaded from http://ahajournals.org by on February 20, 2019
Lindeman and Matsumura Pharmacologic Management of Aneurysms 633
AAA formation in the elastase models follows a typical
pattern with the initial moderate dilatation resulting from loss
of elastic recoil, followed by a secondary dilatation, the ac-
tual aneurysm formation 1 week after the elastase induction
presumably as result of a secondary inflammatory response.
The ultimate dilatation reached varies between 150% and
200%.39 A major criticism of the model is the fact that the
model regresses (heals) and is not associated with rupture,31
although early ruptures were observed after preventive IL
(interleukin)-6 neutralization.40 Other reports show that inter-
ference with the healing response either by TGFβ (transform-
ing growth factor-β) neutralization41 or 3-aminopropionitrile
feeding–induced LOX (lysyl oxidase) inhibition42 elicit rup-
ture in the model.
The second most commonly used model of AAA disease
is generally referred to as the CaCl2 model. In this model,
AAA formation is induced by local calcium salt exposure of
an isolated infrarenal aorta segment.31 Although the model
is scrutinized by some as a minimal model,31 there is a wide
variety in Ca++ concentrations used, and there are indications
that CaPO4 rather than the traditional CaCl2 results in supe-
rior AAA formation.43 Like the traditional elastase model, the
model does not proceed to rupture.
Ruptures form an integral aspect of the third most
commonly used model, the angiotensin II/apolipoprotein
E–deficient mouse.31,32 This model is based on the observa-
tion that chronic angiotensin infusion in apolipoprotein E–
deficient mice results in aneurysms in the aortic tree. Although
the model is commonly referred to as an aneurysm model, it
is now clear that the model should be referred to as a model
of aortic dissection.44,45 Hence, conclusions based on the an-
giotensin model may not or only partially translate to human
AAA disease.
Based on experiments in these 3 models, several hun-
dred targets31 have been proposed to limit aneurysm growth.
Although a detailed review of the interventions is beyond the
scope of this article, successfully targeted main clusters for in-
tervention include vascular inflammation, tissue remodeling,
blood pressure regulation, and lipid metabolism. An overview
of the reported main clusters and illustrative exemplary stud-
ies are provided in Table 1.
The available literature is dominated by positive stud-
ies; few studies report disease aggravation97,98 or failure of
the studied intervention.99–102 With respect to the latter, most
failing interventions are included as a secondary finding pre-
sented along with a successful primary finding, raising the
possibility that the available literature is biased by selective
reporting of positive findings103 and type I errors (false-posi-
tive conclusions). Strong support for the latter stems from an
elegant evaluation by Trachet et al.104 The authors performed
a meta-analysis of the incidence of dissecting aneurysms and
the mortality rates in the control arm of 194 articles report-
ing on the angiotensin model. The analysis revealed strong
inverse relationships between both the aneurysm incidence
and mortality rate of the control group, and the final conclu-
sions of the study (median dissecting AAA incidence: 73%
in studies reporting interventions claimed to reduce AAA
formation, 56% in descriptive studies versus 35% for inter-
ventions claimed to enhance AAA formation).104 Reported
median mortality rates followed the similar inverse trend
(25%, 19%, and 13%, respectively).104
Medical Therapy for Patients With AAA
There are 2 indications for medical therapy in AAA: cardiovas-
cular risk management and pharmaceutical AAA stabilization.
Epidemiological105,106 and cohort studies107–109 character-
ize an AAA as a strong cardiovascular risk factor. In fact,
in patients deemed unfit for repair, the risk of dying from
nonaneurysm-related (in particular, cardiovascular) causes
by far exceeds the risk of dying from AAA.110 The profound
impact of an AAA on overall survival is further illustrated by
the relative survival analysis included in a meta-analysis of
patient survival following open or endovascular repair.111 The
observed 0.76 10-year relative survival ratio for patients who
had their AAA repaired clearly illustrates the profound indi-
rect risk of AAA disease.111
Level IIb evidences suggests that cardiovascular risk man-
agement is effective in patients with AAA112,113; hence there
is a case for cardiovascular risk management for all patients
with AAA, irrespective of a possible impact of the risk man-
agement on aneurysm progression. Logically, improvement
in survival because of reduced cardiovascular risk not only
improves the cost-effectiveness of AAA repair but longer sur-
vival will maximize the benefits of an effective pharmaceuti-
cal stabilization program.
Preclinical models show the potential of lipid lower-
ing,78–82 antihypertensive therapy,72–76 and platelet aggregation
inhibitors59 in quenching experimental AAA development.
Yet, there is little evidence for a beneficial effect of these
strategies on clinical AAA progression and stability. The first
studies exploring the potential of pharmaceutical therapy for
AAA progression were based on observed beneficial associa-
tions between β-blocker use and aneurysm progression in 2
small (n of 12 and 38) case-control studies.114,115 These stud-
ies were then followed by a further case-control116 and cohort
study117 and later by 2 randomized trials.118,119 All these later
studies were not confirmative, although the interpretation of
the randomized controlled trials is compromised by the poor
tolerability of the β-blocker used (propanol), which resulted in
a 42% dropout rate in the treatment arm.119
The ACE (angiotensin-converting enzyme) inhibitors are
the second class of antihypertensive drugs that received signif-
icant attention in the context of AAA stabilization. Enthusiasm
was spurred by supportive evidence from experimental stud-
ies,74,75 and a population-based case-control study that reported
a beneficial association between ACE inhibitor use and risk of
rupture.120 This study was followed by a series of nonconfirma-
tive studies,121–123 one of them suggesting an adverse associa-
tion between ACE inhibitor use and AAA progression.123 These
controversies were ultimately addressed in the AARDVARK
study (Aortic Aneurysmal Regression of Dilation: Value of
Ace-Inhibition on Risk).124 This study concluded that, despite
more effective blood pressure lowering, the ACE inhibitor per-
indopril did not show significant impact on aneurysm growth
(compared with both placebo alone and to combined placebo
and amlodipine [a Ca++ antagonist]). A shortcoming of the
AARDVARK study is the lower-than-anticipated aneurysm
growth. As such, the trial may lack the sensitivity to detect
Downloaded from http://ahajournals.org by on February 20, 2019
634 Circulation Research February 15, 2019
minor effect sizes. Although the authors attribute this short-
coming to the high-level medical cardiac risk management in
the population studied,124 it is likely that the lower-than-antic-
ipated growth reflects inclusion of a disproportionate group of
patients with relatively small AAAs (35 mm).
At this point, the potential of the type 1 angiotensin receptor
antagonist telmisartan is under investigation in the TEDY study
(Effectiveness of Telmisartan in Slowing the Progression of
Abdominal Aortic Aneurysms).125 The rationale for this study is
the fact that AT1 receptor antagonists interfere with the negative
aspects of angiotensin signaling but preserve signaling through
the ATR1 (angiotensin II receptor type 1) receptor, which is
associated with vascular protective activity.126,127 Along these
lines, beneficial associations have been reported between type 1
AT receptor antagonist use and AAA progression.128
The overall conclusion for antihypertensive therapies is
that the available clinical studies refute β-blockers or ACE
inhibitors as pharmaceutical strategies for AAA stabilization.
This indirectly confirms absence of a direct association be-
tween blood pressure and AAA progression.
The potential of statins has been evaluated in 12 studies.
Results of these studies are mixed with 6 studies hinting at a
beneficial association between statin use and AAA progres-
sion129–134 and another 6 studies failing to show an association
between statin use and aneurysm progression.117,122,123,135–137
Conclusions from the studies segregate, with the older and
smaller studies being confirmative and the later and larger
studies being nonconfirmative. On this basis, although the car-
diovascular risk benefits of statins are impressive, there is no
role for statins as a pharmaceutical strategy stabilizing AAA.
An effect of antiplatelet therapy on aneurysm progression
has been explored in 6 studies. Beneficial effects have been ob-
served in medium sized cohort study (n=148) of patients under
surveillance of a 40- to 49-mm AAA.138 Unfortunately, the va-
lidity of the study is challenged by the unrealistic high growth
rate in the control group (5.2 mm/y; anticipated growth rate, 2–
3 mm/y139). A potential effect for combined aspirin-statin treat-
ment has been observed in a subanalysis of a study evaluating
the effect of azithromycin on AAA progression.140 A benefit for
nonsteroidal anti-inflammatory drugs (NSAIDs) has been re-
ported on the basis of a small (n=19) study; reportedly patients
using NSAID showed reduced AAA progression.141
In contrast, 3 larger studies (the UK small aneurysm trial,123
the ADAM study [Aneurysm Detection and Management],117
and an Australian cohort study136) all fail to confirm a benefi-
cial effect of antiplatelet therapy on AAA progression.
Well-established negative (beneficial) associations exist be-
tween diabetic disease and AAA growth rate.142 Although this
has been attributed to diabetes mellitus–related factors, such as
matrix stabilization by enhanced glycation and modulation of in-
flammation,143 there are indications that this negative (protective)
association relates to off-target effects of metformin—a bigu-
anide antidiabetic that is first-line medication for type II diabetes
mellitus. Indeed, metformin use but other classes of antidiabetic
drugs associated with reduced AAA growth rate.144,145 At this
point, 2 trials are planned to test an effect of metformin on AAA
growth (Prof Ron L. Dalman, personal communication).
Above clinical studies, all evaluated potential off-target
(so-called pleiotropic) effects on AAA progression of drugs
that are part of regular cardiovascular risk management. A fur-
ther series of trials evaluate disease-specific targets that were
defined on basis of the current understanding of AAA disease.
A presumed role for persistent chlamydia infection in the per-
petuation of vascular disease, including AAA, at the millennium
Table 1. Summary of Successful Experimental Targets for Pharmaceutical
Abdominal Aortic Aneurysm Stabilization
Targeted Cluster Strategy
Anti-inflammatory NFκB,46 AP-1,47 Rho kinase48 inhibition
IL1,49 TNFα,50 CCL-151
B cell,52 γδT-cell53 depletion
Neutrophil inhibition54
Mast cell inhibition55
Complement inhibition56,57
Oxilipin inhibition58,59
Immune suppression60,61
Protease inhibition MMP inhibition62,63
Cysteine protease inhibition64,65
Serine protease inhibition66,67
Oxidative stress Antioxidant enzymes68,69
Secondary antioxidants70,71
Blood pressure lowering β-Blockers72
Ca antagonists73
ACE inhibitors74,75
ATR1 antagonists76
iNOS inhibition77
Lipid metabolism Statins78,79
HDL80
RXR and PPARα/γ activation81,82
Cell therapy Mesenchymal stem cells83,84
Fibroblasts85
Matrix/morphogens Interference with TGFβ signaling86
Interference with NOTCH87/Wnt88 signaling
Thrombospontin inhibition89
EGFR inhibition90
Metabolism Inhibition of HIF1α91
Activation of AMPK92
Nutriceuticals Polyphenols93
Phytoestrogens94
Sex hormones Castration95
Estrogens96
ACE indicates angiotensin-converting enzyme; AMPK, 5' AMP-activated protein
kinase; AP-1, activator protein-1; ATR1, angiotensin II receptor type 1; CCL-1,
chemokine (C-C motif) ligand; EGFR, epidermal growth factor receptor; HDL, high-
density lipoprotein; HIF1α, hypoxia-inducible factor 1-alpha; IL-1, interleukin-1;
iNOS, inducible NO synthase; MMP, matrix metalloproteinase; NFκB, nuclear
factor-κB; NOTCH, notch homolog, translocation-associated; PPARα/γ,
peroxisome proliferator–activated receptor α/γ; RXR, retinoid X receptor; TGF-β,
transforming growth factor-β; and TNF-α, tumor necrosis factor-α.
Downloaded from http://ahajournals.org by on February 20, 2019
Lindeman and Matsumura Pharmacologic Management of Aneurysms 635
époque resulted in 3 studies, which aimed at anti-chlamydia e-
radication. Two small trials with, respectively, a single 4-week
course of the antibiotic roxithromycin146 or repeated (annual)
4-week courses of roxithromycin147 reported borderline benefits
on aneurysm progression. However, this was not confirmed in a
larger study with azithromycin that did not identify an effect of
16 weeks of macrolide treatment on AAA progression.140
A further series of clinical studies aimed at targeting spe-
cific aspects of the vascular inflammation and proteolytic im-
balance in AAA. In this respect, there is a longstanding interest
in the tetracycline antibiotic doxycycline. Independent of its an-
tibiotic properties, doxycycline has been shown to reduce the
expression of matrix metalloproteinases148 and to quench their
activity.149 Doxycycline effectively interferes with aneurysm
formation in some150,151 but not all99 models of aneurysm for-
mation, and clinical studies showed that doxycycline treatment
reduces aortic wall MMP (matrix metalloproteinase) content
and improves the proteolytic imbalance through its effect on
aneurysm wall protease inhibitor levels.152,153 Three studies154–156
evaluated the effect of doxycycline treatment on aneurysm pro-
gression, and a fourth multicenter randomized trial N-TA^3CT
(Noninvasive Treatment of Abdominal Aortic Aneurysm
Clinical Trial) is ongoing.157 A first small study (n=32) evalu-
ated patients after 3 months of doxycycline eradication therapy
with the intent of testing the persistent chlamydia infection hy-
pothesis.154 The report claims an effect of doxycycline therapy
on aneurysm progression in the 6- to 12- and 12- to 18-month
follow-up intervals, but no effect was seen for the initial 0- to
6-month interval and for the overall study period. The second
open phase II study tested the safety and feasibility of 6 months
of doxycycline therapy in patients with AAA.155 The study
showed that chronic doxycycline treatment is feasible and well
tolerated, and it was concluded that aortic wall MMP-9 levels
and AAA progression compared favorably with those of his-
toric controls not receiving treatment. In contrast, data from the
PHAST trial (Pharmaceutical Aneurysm Stabilization Trial)156
testing the effect of 18 months of doxycycline (100 mg/day)
failed to show a benefit of doxycycline therapy on AAA pro-
gression; on the contrary, doxycycline treatment resulted in a
statistically significant but clinically insignificant acceleration
of AAA growth. Although earlier dose-finding study found dose
equivalence for low-, regular-, and high-dose (50, 100, or 300
mg/day, respectively) doxycycline on all parameters tested,158,159
it has been argued that the dose used in the PHAST study is too
low to elicit an effect. As such, the results of the N-TA^3CT
study that tests the benefit of 200 mg/day157 are eagerly awaited.
A potential benefit of mast cell inhibition through the potent
mast cell stabilizer pemirolast160 was tested in the AORTA trial
(CRD007 for the Treatment of Abdominal Aorta Aneurysm).161
Study results show that 12 months of mast cell inhibition is safe,
but the mast cell inhibition did not influence AAA progression.161
The ACZ885 (canakinumab) for the Treatment of
Abdominal Aortic Aneurysm study162 tested the effect of IL-
1β neutralization through subcutaneous canakinumab (150
mg) once per month for 12 months. The study enrolled 65 pa-
tients, and 1 year growth data was obtained for 20 participants
in the placebo group and 23 in the canakinumab group. AAA
progression (2.5 mm/y) was similar for both groups, and the
trial was terminated for reasons of futility.162
Apart from these studies on diameter changes, there are
data available for surrogate end points (aneurysm wall inflam-
mation). In a small study (6 cases, 10 controls), Motoki et
al163 evaluated the effect of PPAR (peroxisome proliferator–
activated receptor)-γ agonist pioglitzone and observed a re-
duction in aortic wall TNF-α (tumor necrosis factor-α) and
MMP-9 expression. The effects of the PPARα agonist fe-
nofibrate on circulating inflammatory markers of inflamma-
tion have been studied in the FAME trial (Fenofibrate in the
Management of Abdominal Aortic Aneurysm). A randomized
study of 24 weeks of fenofibrate treatment in AAA patients.164
No effect was observed on the circulating markers osteopontin
or kallistatin. Although the authors report an absent effect on
AAA growth,164 it is important to point out that the trial was
not adequately powered to detect such an effect.
A highly specific suppression of aneurysm wall inflam-
mation was observed for the selective vitamin D receptor ago-
nist paricalcitol. It was shown that a 2- to 4-week preoperative
paricalcitol treatment selectively interfered with aspects of
NFAT2 (nuclear factor of activated T cells-2) mediated inflam-
mation,165 suggesting that the effects of vitamin D are mainly
mediated by an effect on calcineurin-mediated inflammation.
This notion was confirmed in in vitro studies.165 Although
plasma lipids do not associate with incident AAA disease,
there are weak associations between plasma LDL (low-den-
sity lipoprotein) levels and AAA progression.166 In this light,
the observed superior effects of ezetimide/simvastatin over
simvastatin alone on vascular inflammation merit attention,167
yet it is unclear how these observations relate to the apparent
absence of statins on AAA progression.117,122,123,135–137
The above overview of preclinical successes and clinical
failures points to a major paradox of numerous preclinical
successes and clinical challenges. Preclinical studies identi-
fied hundreds of successful candidate interventions, yet this
enormous investment has not produced any clinical applica-
tion, and no medical therapy is currently available for the sta-
bilization of growing AAA, although further clinical studies
are underway or planned (Table 2).
More important, the apparent translational gap between
preclinical and clinical studies challenges our concepts of the
processes underlying late-stage AAA disease pathophysiol-
ogy. Undoubtedly, AAA is associated with a sustained and
comprehensive inflammatory response, uncontrolled protease
activity, and excess matrix turnover.5,20,153 Short-term preopera-
tive intervention studies in patients undergoing open repair all
proved the potential of indomethacin,168 statins,137,169–176 ACE
inhibitors,121 and doxycycline153,158,159 to effectively quench
vascular inflammation and protease activity. Yet, these effects
are not followed by reduction of AAA progression. Along
similar lines, bona fide anti-inflammatory strategies, such as
anti–IL-1β therapy162 and mast cell161 stabilization failed to
influence AAA progression. Interestingly, profound immune
suppression in the context of solid organ transplantation even
results in accelerated AAA progression.177,178 Although these
aforementioned observations do not exclude a role for inflam-
mation or protease activity in AAA initiation and progression,
they imply involvement of additional, to date unidentified crit-
ical factors that are unresponsive to the anti-inflammatory/an-
tiproteolytic therapies.
Downloaded from http://ahajournals.org by on February 20, 2019
636 Circulation Research February 15, 2019
One of the possible key factors is failing or defective
compensatory repair. In fact, interference with compensatory
repair mechanisms (stem cell function) may explain the ap-
parent disastrous effects of intense immune suppression,179
chemotherapy,180 and the unexpected negative effect of dox-
ycycline therapy156,181 on aneurysm growth. Moreover, there
are clear indications for defective matrix repair in AAA. The
disease is associated with complete loss of the normal aortic
wall architecture, and the normal aortic matrix is replaced by
a collagenous, fibrotic matrix.153 Although a higher collagen
cross-link content in AAA wall samples may imply more sta-
ble collagen,182 this is actually not the case because of defects
at the level of collagen fibril organization. In the healthy aortic
matrix, the collagen fibrils are laid down in supramolecular, in-
tertwined network structures. As a result, forces are distributed
over the wall. Loss of this network behavior in AAA disease
fundamentally impacts the mechanical stability of the wall182
and may contribute to the aortic wall weakening in the disease.
Fatty degeneration was recently identified as another poten-
tial contributor to the weakening of the aneurysm wall.183,184 Fatty
degeneration is a known phenomenon in aging and chronically
injured muscle185 and is thought to be a consequence of impaired
repair mechanism in the context of chronic injury.186 Gene-
expression studies on AAA wall specimens suggest that progres-
sive adipocyte accumulation associated with rupture.183,187
Unfortunately, the perpetual inflammatory cycle and the im-
paired compensatory repair that hallmark human AAA are not
captured in the rodent models of AAA disease. This shortcoming
may largely reflect the spontaneous resolution of inflammation
in these models and the superior endogenous healing responses
of small animal models,188,189 as well as their inherent resistance
to develop chronic fibrosis. In an attempt to create more relevant
(viz rupture prone) AAA models, modified models have been
introduced in which interference with the primary healing re-
sponses resulted in AAA ruptures.41,42 Yet, these models do not
recapitulate the chronically impaired and dysregulated healing
responses that characterize AAA disease. Absence of fibrotic re-
pair in murine models of AAA disease also explains the apparent
benefit of inducing fibrotic repair in stabilizing growing AAA
in murine models.190 Because extensive fibrosis is a hallmark of
human AAA disease, and that process of fibrosis results in dep-
osition of a brittle, poor-quality matrix,153,182 it is questionable
whether a profibrotic strategy will stabilize human AAA.
Considering the wealth of preclinical success and failing clini-
cal attempts to identify molecular strategies for stabilizing AAA
disease, we must acknowledge that our understanding of AAA
disease is far from complete and that the available small animal
models of the disease only partially mimic aspects of the human
disease. There appears a recent trend to include (or demand)
confirmative studies in a second animal model in preclinical
studies. Considering the parallels between the different models,
it is dubious whether this increases the likelihood of the find-
ings being more translationally relevant. Future advancement
of the field critically relies on an improved mechanistic insight
in the processes that sustain the impaired and ultimately failing
repair mechanisms in advanced clinical AAA disease.
Table 2. Ongoing and Planned Medical Intervention Studies for AAA Stabilization
Study Acronym Intervention Readout
N-TA^3CT,157 NCT01756833 Doxycycline 100 mg BID or placebo 2-y AAA progression (CT scan), repair,
or rupture
TEDY,125 NCT01683084 Telmisartan (AT1 receptor antagonist) 40 mg or placebo 1-y AAA progression (ultrasound and CT
scan), repair, or rupture
VIVAAA, NCT02846883 Mesenchymal stem cells AAA inflammation (PET-CT)
Eplerenone in the Management of Abdominal Aortic
Aneurysms, NCT02345590
Eplerenone (selective aldosterone receptor antagonist) or
placebo
Not specified
TicAAA, NCT02070653 Placebo or none specified dose ticagrelor (P2Y12 receptor
inhibitor)
1-y AAA progression, surgery, or repair.
FAME,191
ACTRN12612001226897
Fenofibrate (PPARα agonist) 145 mg or placebo 2–4 wk
before elective open repair
Aortic wall macrophage and osteopontin
content
The Effect of Angiotensin II Type 1 Receptor Antagonists
on the Size and Expansion Rate of Abdominal Aortas in
Hypertensive Patients, NCT01670903
Comparison of patients treated with different classes of
antihypertensives (AT1 receptor antagonists, (ARB) ACE
inhibitors, or non-ARB/ACE)
Not specified
Metformin Therapy in Nondiabetic AAA Patients, NCT03507413 Metformin (1000 mg BID) or placebo 1-y AAA progression (CT)
LIMIT trial, not yet registered Metformin or placebo 2-y AAA progression (CTA)
IMAGEN192 Inositol or placebo 1-y AAA progression (sack volume [CT])
ARREST trial193 1 or 3 106 cells/kg allogenic mesenchymal
cells or placebo
Phase I safety trial. Circulating cytokine
levels and 18-FDG/PET
AAA indicates abdominal aortic aneurysm; ACE, angiotensin-converting enzyme; ARREST, Aortic Aneurysm Repression With Mesenchymal Stem Cells; CT, computed
tomography; CTA, computed tomography angiography; FAME, Fenofibrate in the Management of Abdominal Aortic Aneurysm; FDG, fludeoxyglucose; IMAGEN, Inositol in
the Management of Abdominal Aortic Aneurysm; LIMIT, Limiting AAA With Metformin; N-TA^3CT, Noninvasive Treatment of Abdominal Aortic Aneurysm Clinical Trial;
PET, positron emission tomography; PPARα, peroxisome proliferator–activated receptor-α; TEDY, Study of the Effectiveness of Telmisartan in Slowing the Progression
of Abdominal Aortic Aneurysms; TicAAA, The Efficacy of Ticagrelor on Abdominal Aortic Aneurysm Expansion; and VIVAAA, Safety and Efficacy of Allogeneic MSCs in
Promoting T-Regulatory Cells in Patients With Small Abdominal Aortic Aneurysms.
Downloaded from http://ahajournals.org by on February 20, 2019
Lindeman and Matsumura Pharmacologic Management of Aneurysms 637
Marfan Syndrome
Marfan syndrome is an autosomal-dominant, multisystem con-
nective tissue disorder. The syndrome is caused by mutations
in the Fbn-1 (fibrillin-1) gene region located on chromosome
15 and is estimated to affect 2 to 3 of 10 000 individuals.
Over 1000 different Fbn1 mutations have been associated
with the syndrome,194 and the syndrome has extreme heter-
ogeneous genotype-phenotype variability195 (see Table 3 for
the diagnostic criteria196). Ascending, and to a slighter lesser
extent descending, thoracic aortic aneurysms are among the
primary disquieting features of the syndrome.197,198
The Fbn-1 gene codes for fibrillin—a structural connective
tissue macromolecule that has been traditionally been consid-
ered a key chaperone in elastic fiber formation. However, in-
volvement of tissues not containing elastin indicates roles far
beyond that as a scaffold of elastin formation. Indeed, defects
in the Fbn-1 gene associate with impaired collagen network
formation,181 and fibrillin is a complex modulator of growth
factor signaling and cell function.199
The aortapathy (thoracic aneurysms, dissections) is among
the leading causes of premature death in patients with Marfan.
Although increased awareness, improved surgical techniques,
and medical therapy have significantly improved prospects,
Marfan syndrome still comes with significant aorta-related
mortality.200,201 In this respect, strong associations have been
described between the gross genotype (dominant-negative
[abnormal FBN1 protein] or haploinsufficient [reduced FBN1
protein]) and survival, with significant better outcomes in pa-
tients with dominant-negative mutations.202,203
Aneurysms in Marfan syndrome are currently managed
by medical therapy (β-blockers and possibly ATR1 antago-
nists [AT2 inhibitors]) and preventive surgical repair once the
aneurysm size exceeds 50 mm.204 Although medical therapy
preventing aortic dilatation has a prominent role in the current
guidelines, it is important to note that the level of evidence is
low.
Recommendations for β-blocker therapy are actually
based on a single, small open study that included 70 patients.205
Conclusions from this study have recently been scrutinized
on basis of its small size and considerable losses during fol-
low-up, and the fact that significance was only reached on
creating a composite end point.206 In addition to this single
intervention study, there is supportive data from a series of
observational reports.
The largest observational study is by Silverman et al.207
The authors reported outcomes for 417 patients with definite
Marfan syndrome who were under surveillance in 4 referral
centers. Groups were created on basis of β-blocker prescrip-
tion. β-blocker usage was unknown for 84 patients, as a re-
sult, 191 patients taking β-blockers and 142 patients who had
never taken β-blockers were evaluated. Despite the impressive
study size, this study comes with significant points of concern.
According to the authors, “Median cumulative probability of
survival for patients who had taken β-blockers was 72 years
compared with 70 years for patients who had never taken β-
blockers (P=0.01).” Yet, the reported estimated life expectan-
cies contrast with actual data in the article and with the data
for other populations for the same time interval.200 Moreover,
it is important to point out that the number of patients with
an age >50 years in the study was limited,207 and as a conse-
quence that the study is underpowered to allow detection of a
2-year difference in life expectancy. A further issue with the
study is the fact that authors did not address putative time ef-
fects in their analysis. Although not fully clear from the text,
it appears that the study covers the period between 1970
and 1993. It is conceivable that improvements in surgical
techniques coincided with the clinical implementation of β-
blocker usage; making time is a major potential confounder in
this study. As actually pointed out by the authors in the discus-
sion, “it is very likely that increased awareness and improved
diagnostic tools resulted in progressively more mild cases of
the Marfan syndrome being identified towards the end of the
observation period.207
A further observational study208 on an effect of β-blockers
on aortapathy reports growth data for 113 juvenile patients
with Marfan from 2 centers. Different dosing schedules were
used by the 2 centers: an intermediate dose in the first cen-
ter (1.3 mg/kg; n=80 patients) and a high dose in the second
center (1.9 mg/kg; n=20 patients). Thirteen individuals who
could not or would not take β-adrenergic blockade therapy
constituted the control group.208 On the basis of the slower
rate of aortic root growth in individuals taking β-blockers, the
authors recommended that “β-adrenergic blockade therapy in
patients with Marfan syndrome should begin at the earliest
age possible, and that the dose be adjusted to the largest dose
β-adrenergic blockade therapy that is clinically tolerated.208
Some concerns of this study include the authors report benefi-
cial effects on the aorta growth rate; yet, this is actually not the
case for the indexed growth rate (mm/m2), for which favorable
effects were only observed for the intermediate-dose group
and not for high-dose group. Reviewing the article208 for a
Table 3. Revised Ghent Criteria for Diagnosing Marfan Syndrome*
In the absence of a family history
1. Z score for the aortic diameter at the sinuses of valva 2 or aortic root
dissection and ectopia lentis
2. Z score for the aortic diameter at the sinuses of valva 2 or aortic root
dissection and FBN1 mutation
3. Z score for the aortic diameter at the sinuses of valva 2 or aortic root
dissection and systemic score 7*
4. Ectopia lentis and FBN1 mutation and aortic aneurysm
In the presence of a family history
5. Ectopia lentis and family history of Marfan syndrome (see 1–4)
6. Systemic score 7 and family history of Marfan syndrome (see 1–4)*
7. Z score for the aortic diameter at the sinuses of valva 2 (>20 y old)
or 3 in those <20 y old) and family history of Marfan syndrome*
Systemic score—Wrist and thumb sign: +3 (wrist or thumb sign: +1); pectus
carinatum deformity: +2 (pectus excavatum or chest asymmetry: +1); hindfoot
deformity: +2 (plain pes planus: +1); pneumothorax: +2; dural ectasia: +2;
protrusio acetabuli: +2; reduced upper/lower segment and increased arm/height
and no severe scoliosis: +1; scoliosis or thoracolumbar kyphosis: +1; reduced
elbow extension: +1; facial features (3/5): +1 (dolichocephaly, enophtalmos,
downslanting palpebral fissures, malar hyoplasia, and retrognathia); skin striae:
+1; myopia >3 diopters: +1; mitral valve prolapse (all types): +1. Maximum total:
20 points; score 7 indicates systemic involvement. FBN1 indicates fibrillin-1.
*In the absence of discriminating features other genetic syndromes.
Downloaded from http://ahajournals.org by on February 20, 2019
638 Circulation Research February 15, 2019
potential explanation(s) reveals that with similar mean end-of-
follow-up ages in the intermediate-dose and control groups,
mean end-of-follow-up length in the intermediate-dose group
was 174 cm but only 149 cm in the control group.208 An ex-
treme SD in the control group (69 cm [versus 22 cm in the
intermediate-dose group])208 implies severe skewing of the
size distribution to the right in the control group and conse-
quently that the reported mean height overestimates the actual
median height. This implies profound heterogeneity between
control group and the treated groups and consequently that the
conclusions of the study may be prone to bias.
Beneficial effects are further reported by Ladouceur et
al,209 who retrospectively evaluated the effect of β-blockers
in 155 young patients with Marfan in whom the therapy was
initiated before the age of 12 years. The authors concluded
that “β-blockade significantly decreased the rate of aortic
dilatation at the level of the sinuses of Valsalva by a mean
of 0.16 mm/y (P<0.05), an effect that increased with treat-
ment duration.”209 Although the authors rightly point out that
the increase in aortic dilatation was less in the treatment arm,
this difference actually reflects the larger baseline diameter
in group receiving β-blockers because the actual aortic di-
ameters at the age of 18 years were actually similar in the 2
groups. The claim made by the authors that “a trend toward
lower cardiac mortality, decreased need for preventive aortic
surgery, and less dissection was observed”209 is not justified by
the data in the article.
Conclusions from Ladouceur et al209 are not confirmed in
a second smaller observational study in young patients with
Marfan.210 This study included 63 children who were moni-
tored for >6 years. Thirty-four patients received β-adrenergic
blockade therapy (atenolol, 0.92 mg/kg), 29 patients not re-
ceiving β-blockers served as control. The authors concluded
that “This study found no difference in the rate of aortic root
dilation in children with Marfan syndrome treated with β-
blockers and those not treated.210 Like the other reports, this
retrospective analysis is prone to bias. In particular, a higher
percentage of patients with a family history of Marfan syn-
drome in the untreated group (35% versus 69% in the treated
group) may indicate that groups were not balanced with re-
spect to the severity or phenotype.
A further small, open-label study211 nonrandomly assigned
58 adolescent patients with Marfan to β-blocker therapy
(maximum dose, 2 mg/kg) or the ACE inhibitor enalapril. It
was concluded that ACE inhibition resulted in favorable he-
modynamic changes and a smaller increase in aortic root di-
ameter (0.1 [1.0] versus 5.8 [5.2] mm [mean (SD)]).211 Given
the study design, the small sample size, and absence of a con-
trol group, it is difficult to draw conclusions from this study.
A report from Rossi-Foulkes et al212 compares outcomes for
preadolescent patients on different antihypertensive therapy
(β-blockers or Ca antagonists). The authors reported that med-
ication favorably influenced aortic growth,212 but that it did not
prevent complications. In the absence of a control group, and
profound baseline differences in the medicated and nonmedi-
cated group, this report should be considered inconclusive.
Taken together, this overview of reports on β-adrenergic
blockade in Marfan syndrome shows a paucity of stud-
ies with adequate study designs and appropriate statistical
approaches.213 As a consequence, the currently available ev-
idence does not provide a strong rationale for β-adrenergic
blockade to prevent aortapathy in patients with Marfan syn-
drome.214–216 An adequate evaluation taking into account the
possibility that patient responses to β-adrenergic blockade are
heterogeneous and relate to the underlying genotype217,218 is
missing.
Observed excessive TGFβ signaling in the aortas of murine
models of Marfan syndrome and a preventive effect of inter-
ference with TGFβ signaling through neutralizing antibodies
or the angiotensin II receptor antagonists in the model219 fu-
eled optimism for angiotensin II receptor antagonists (sartans)
as a preventive treatment for aortapathy in Marfan syndrome.
Supportive observations from small (28, 20, and 18 pa-
tients, respectively) open studies in young patients with
Marfan220–222 and a small open-label study on surrogate end
points223 were followed by 1 smaller and 4 larger randomized
trials. A small Belgium trial enrolling 22 patients with Marfan
syndrome failed to observe an add-on effect of losartan when
added to blocker therapy.224
Forteza et al225 performed a larger randomized trial and
randomized 5- to 60-year-old Marfan patients to losartan
(n=70) or atenolol (n=70; both dosed at 100 mg/day in indi-
viduals >50 kg). The trial results show similar aortic root and
ascending aortic diameter progression in the 2 arms for the
3-year follow-up.225 In a French study, incorporating 303 pa-
tients with Marfan aged 10 years, Millerton et al226 assigned
patients to losartan or placebo next to their regular treatment
(86% of the participants also used β-blockers). It was con-
cluded that 3-year losartan therapy did not influence the aorta
parameters tested or the need for surgery.226 Unfortunately, in-
clusion of both young and adult patients creates considerable
heterogeneity both with regard to the geno/phenotype as to
aortic dilation rates potentially interfering with the ability to
detect suppression of growth.
Young patients with Marfan were studied in a semi-blinded
study by the US Marfan network. Six hundred and eight par-
ticipants between 6 months to 25 years of age were allocated
to atenolol (mean dose [SD] achieved, 2.7 [1.1] mg) or losartan
(mean dose [SD], 1.3 [0.2] mg).227 Again, the 3-year follow-up
showed equivalence for β-adrenergic blockade or angiotensin
receptor blockade. The fourth larger randomized trial is a mul-
ticentre, open-label, randomized controlled trial with blinded
assessments performed in the Netherlands.228 The COMPARE
trial (Cozzar in Marfan Patients Reduces Aortic Enlargement)
incorporates 233 adult participants (47% women) who were
randomized to either losartan (n=116) or no additional treat-
ment (n=117). The study showed mixed effects with an effect
of losartan on root dilatation rate but no effect on the more dis-
tal aspects of the aorta. Remarkably, a planned subanalysis per-
formed on the available data of the COMPARE trial228 suggests
that an effect of angiotensin II receptor blockade may depend
on the type of FBN1 mutation because it was concluded that
losartan reduced only aortic root dilatation rate in haploinsuf-
ficient patients and not in dominant-negative patients.229
According to the trial registries, there is currently 1 small
(n=56) ongoing 4-arm trial, The Oxford Marfan Trial version,
which evaluates the effect of irbesartan (150–300 mg), doxy-
cycline (100–200 mg), and a combination of both on markers
Downloaded from http://ahajournals.org by on February 20, 2019
Lindeman and Matsumura Pharmacologic Management of Aneurysms 639
of vascular dysfunction in the Marfan syndrome in patients
>13 years of age.230
With the exception of the potential beneficial effect in
adult haploinsufficient Marfan patients, the clinical trials
uniformly fail to show a benefit of type I angiotensin II re-
ceptor inhibition on the aortapathy in patients with Marfan.
Remarkably, opposite conclusions were drawn in a meta-
analysis of the published prospective trials.231 Evaluation of
the meta-analysis appears to have weaknesses that resulted in
an overestimation of the effect size. Specifically, the planned
subanalyses performed within the COMPARE trial228 were
included as separate studies, resulting in duplication of the
positive data. Further, the weight distribution attributed to the
studies included in the meta-analysis231 may be incorrect.
These contrasting findings between promising preclinical
data and the clinical data with respect to the angiotensin II
receptor antagonism may reflect profound interspecies differ-
ences, not only with respect to aspects of the immune and in-
flammatory responses but also with respect to healing, as well
as the significant heterogeneous character of aorta disease in
Marfan disease.232 Most of the preclinical work is based on
mice with hypomorphic FBN1 mutations (the Fbn1C1039G/+
strain, with 50% of normal and Fbn1mgR/mgR strain with
20% of normal FBN1),233 but alternative models are currently
being developed.234
Given the extreme genotypic and phenotypic varia-
tion in Marfan syndrome, observations from a specific mu-
rine model may only be relevant to a subset of patients with
Marfan. Translatability of experimental findings can be fur-
ther interfered by phenotypic aspects such age (disease stage).
Heterogeneity is indicated by the dimorphic effects of TGFβ
neutralization in an experimental model of Marfan syn-
drome.235 Clinical relevance of the genotypic heterogeneity is
as implicated in the subanalysis of the COMPARE trial that
showed an exclusive benefit of losartan in haploinsufficient
patients.229 As such, a reevaluation of the negative trials tak-
ing along the lines of dominant-negative and haploinsufficient
genotypes merits consideration.
Unfortunately, such a meta-analysis may be challenging
because phenotyping was only available for one-third of the
patients in the large Atenolol Versus Losartan Trial performed
by the Pediatric Heart Network.227 Genotype information is
available for 84% of the participants in the Sartan trial (78%
established FBN1 mutation)226 and the trial by Forteza et al225
(82% FBN1 mutation), but information on type FBN1 is miss-
ing in the publications.
Interpretation of currently available randomized trials is
further challenged by the substantial phenotypic heterogene-
ity in the patients studied (young versus adult patients) and by
loss of sensitivity by use of Z scores rather than root size as
clinical end points.236 A meta-analysis based on all individual
patient data has been announced, but conclusions are awaited
at the time of writing this overview.237
A further point of debate is the suggested pivotal role of
TGF-β signaling in Marfan disease.219 The rationale for ATR1
blocker in Marfan syndrome was based on a presumed ex-
cess TGF-β signaling as the underlying cause of aortapathy in
Marfan syndrome. A critical question is whether this assump-
tion is correct as excess TGF-β activation appears a common
phenomenon in aortic aneurysm disease238,239 and may actu-
ally be part of the compensatory healing or anti-inflammatory
responses. Such a mechanism is supported by the observation
that TGF-β upregulation in murine models of Marfan syn-
drome is secondary,238,240–242 by the fatal consequences of TGF-
β neutralizing in murine models of aneurysm41,243 and opposite
contextual (disease state) effects of TGFβ on the aorta pathol-
ogy in the Marfan mouse model.235
Taking into account the currently available data, there is in-
sufficient evidence in support for either β-adrenergic blockade
or ATR1 blocker for aortapathy in Marfan syndrome. The field
would benefit from the announced meta-analysis (and subanaly-
sis) of the available data from the losartan trials.237 If this is not
definitive, there is need for an adequately powered, placebo-con-
trolled global trial that would stratify or control multiple known
confounders, including age, genetic heterogeneity of Marfan
syndrome, and diverging effects on blood pressure and pulse.
Currently available data indicate equivalence of β-
adrenergic and ATR1 blockade. In light of the milder side ef-
fects and superior persistence,244 ATR1 antagonists might be
preferable.245
Summary
The inconsistent correlations of preclinical successes and
clinical trial results provide impetus for major advances in re-
search innovation of aortic aneurysm. There are several expla-
nations for this translational gap, including inadequate animal
models, incomplete understanding of the processes involved
in the progression of late stage human disease, poorly de-
signed, underpowered clinical trials, and selective reporting.
Development of humanized models, advancing beyond early
chemically provoked models to those consistently demonstrat-
ing sequential dilation and rupture, addressing dysfunctional
repair mechanisms, clinical studies utilizing digitized phar-
maceutical histories and extensive prior radiographic records
of aneurysm size to explore relationships without the heavy
expense of prospective trials, imaging and genetic biomarkers
predicting stability or rapid progression of aneurysm enlarge-
ment, run-in periods ensuring populations with active aortic
enlargement before randomization of subjects, greater atten-
tion to concomitant medical therapy of cardiovascular risk
factors, and longer follow-up with clinical end points in clin-
ical trials are all areas of opportunity in this important field.
Sources of Funding
This study was supported by research grant funding to Leiden
University Medical Center (J.H. Lindeman): Abvie, Cardoz, Eli Lilly,
The Netherlands Organisation for Health Research and Development,
the Dutch Heart Foundation, and the NutsOhra Fund and research
grant funding to the University of Wisconsin (J.S. Matsumura):
Abbott, Gore, Endologix, Cook, Medtronic, and National Institutes
of Health.
Disclosures
None.
References
1. Tedesco MM, Dalman RL. Arterial aneurysms. In: Cronenwett J, Johnston
KW, eds. Rutherfords Vascular Surgery. Philadelphia, PA: Saunders
Elsevier; 2010:117–130.
Downloaded from http://ahajournals.org by on February 20, 2019
640 Circulation Research February 15, 2019
2. Pomposelli FB, Hamdan A. Lower extremity aneurysms. In: Cronenwett
J, Johnston KW, eds. Rutherfords Vascular Surgery. Philadelphia, PA:
Saunders Elsevier; 2010:2110–2127.
3. Roche-Nagle G, Wooster D, Oreopoulos G. Popliteal vein aneurysm. Am J
Surg. 2010;199:e5–e6. doi: 10.1016/j.amjsurg.2009.03.023
4. Meester JAN, Verstraeten A, Schepers D, Alaerts M, Van Laer L,
Loeys BL. Differences in manifestations of Marfan syndrome, Ehlers-
Danlos syndrome, and Loeys-Dietz syndrome. Ann Cardiothorac Surg.
2017;6:582–594. doi: 10.21037/acs.2017.11.03
5. Wågsäter D, Paloschi V, Hanemaaijer R, Hultenby K, Bank RA, Franco-
Cereceda A, Lindeman JH, Eriksson P. Impaired collagen biosynthesis and
cross-linking in aorta of patients with bicuspid aortic valve. J Am Heart
Assoc. 2013;2:e000034. doi: 10.1161/JAHA.112.000034
6. Grewal N, Gittenberger-de Groot AC, Poelmann RE, Klautz RJ, Lindeman
JH, Goumans MJ, Palmen M, Mohamed SA, Sievers HH, Bogers AJ,
DeRuiter MC. Ascending aorta dilation in association with bicuspid aortic
valve: a maturation defect of the aortic wall. J Thorac Cardiovasc Surg.
2014;148:1583–1590. doi: 10.1016/j.jtcvs.2014.01.027
7. Abdul-Hussien H, Soekhoe RG, Weber E, von der Thüsen JH, Kleemann
R, Mulder A, van Bockel JH, Hanemaaijer R, Lindeman JH. Collagen deg-
radation in the abdominal aneurysm: a conspiracy of matrix metallopro-
teinase and cysteine collagenases. Am J Pathol. 2007;170:809–817. doi:
10.2353/ajpath.2007.060522
8. Miller DV, Maleszewski JJ. The pathology of large-vessel vasculitides.
Clin Exp Rheumatol. 2011;29:S92–S98.
9. Ruddy JM, Jones JA, Ikonomidis JS. Pathophysiology of thoracic aortic
aneurysm (TAA): is it not one uniform aorta? Role of embryologic origin.
Prog Cardiovasc Dis. 2013;56:68–73. doi: 10.1016/j.pcad.2013.04.002
10. Molema G. Heterogeneity in endothelial responsiveness to cytokines,
molecular causes, and pharmacological consequences. Semin Thromb
Hemost. 2010;36:246–264. doi: 10.1055/s-0030-1253448
11. Topouzis S, Majesky MW. Smooth muscle lineage diversity in the chick
embryo. Two types of aortic smooth muscle cell differ in growth and re-
ceptor-mediated transcriptional responses to transforming growth factor-
beta. Dev Biol. 1996;178:430–445. doi: 10.1006/dbio.1996.0229
12. Sinha S, Iyer D, Granata A. Embryonic origins of human vascular smooth
muscle cells: implications for in vitro modeling and clinical application.
Cell Mol Life Sci. 2014;71:2271–2288. doi: 10.1007/s00018-013-1554-3
13. Leroux-Berger M, Queguiner I, Maciel TT, Ho A, Relaix F, Kempf H.
Pathologic calcification of adult vascular smooth muscle cells dif-
fers on their crest or mesodermal embryonic origin. J Bone Miner Res.
2011;26:1543–1553. doi: 10.1002/jbmr.382
14. Trigueros-Motos L, González-Granado JM, Cheung C, Fernández
P, Sánchez-Cabo F, Dopazo A, Sinha S, Andrés V. Embryological-
origin-dependent differences in homeobox expression in adult a-
orta: role in regional phenotypic variability and regulation of
NF-κB activity. Arterioscler Thromb Vasc Biol. 2013;33:1248–1256. doi:
10.1161/ATVBAHA.112.300539
15. Pryshchep O, Ma-Krupa W, Younge BR, Goronzy JJ, Weyand
CM. Vessel-specific Toll-like receptor profiles in human me-
dium and large arteries. Circulation. 2008;118:1276–1284. doi:
10.1161/CIRCULATIONAHA.108.789172
16. Stather PW, Sidloff DA, Rhema IA, Choke E, Bown MJ, Sayers RD. A
review of current reporting of abdominal aortic aneurysm mortality and
prevalence in the literature. Eur J Vasc Endovasc Surg. 2014;47:240–242.
doi: 10.1016/j.ejvs.2013.11.007
17. Jones GT, Tromp G, Kuivaniemi H, et al. Meta-analysis of genome-
wide association studies for abdominal aortic aneurysm identifies
four new disease-specific risk loci. Circ Res. 2017;120:341–353. doi:
10.1161/CIRCRESAHA.116.308765
18. Bradley DT, Badger SA, McFarland M, Hughes AE. Abdominal aor-
tic aneurysm genetic associations: mostly false? A systematic review
and meta-analysis. Eur J Vasc Endovasc Surg. 2016;51:64–75. doi:
10.1016/j.ejvs.2015.09.006
19. Moll FL, Powell JT, Fraedrich G, Verzini F, Haulon S, Waltham M, van
Herwaarden JA, Holt PJ, van Keulen JW, Rantner B, Schlösser FJ, Setacci
F, Ricco JB; European Society for Vascular Surgery. Management of ab-
dominal aortic aneurysms clinical practice guidelines of the European
society for vascular surgery. Eur J Vasc Endovasc Surg. 2011;41(suppl
1):S1–S58. doi: 10.1016/j.ejvs.2010.09.011
20. Thompson RW, Geraghty PJ, Lee JK. Abdominal aortic aneurysms: basic
mechanisms and clinical implications. Curr Probl Surg. 2002;39:110–230.
21. Brewster DC, Cronenwett JL, Hallett JW Jr, Johnston KW, Krupski WC,
Matsumura JS; Joint Council of the American Association for Vascular
Surgery and Society for Vascular Surgery. Guidelines for the treatment of
abdominal aortic aneurysms. Report of a subcommittee of the Joint Council
of the American Association for Vascular Surgery and Society for Vascular
Surgery. J Vasc Surg. 2003;37:1106–1117. doi: 10.1067/mva.2003.363
22. Tomee SM, Lijftogt N, Vahl A, Hamming JF, Lindeman JHN. A registry-
based rationale for discrete intervention thresholds for open and endovas-
cular elective abdominal aortic aneurysm repair in female patients. J Vasc
Surg. 2018;67:735–739. doi: 10.1016/j.jvs.2017.07.123
23. Egorova NN, Vouyouka AG, McKinsey JF, Faries PL, Kent KC,
Moskowitz AJ, Gelijns A. Effect of gender on long-term survival after
abdominal aortic aneurysm repair based on results from the Medicare
national database. J Vasc Surg. 2011;54:1–12.e6; discussion 11. doi:
10.1016/j.jvs.2010.12.049
24. Epstein D, Sculpher MJ, Powell JT, Thompson SG, Brown LC, Greenhalgh
RM. Long-term cost-effectiveness analysis of endovascular versus open
repair for abdominal aortic aneurysm based on four randomized clinical
trials. Br J Surg. 2014;101:623–631. doi: 10.1002/bjs.9464
25. Paravastu SC, Jayarajasingam R, Cottam R, Palfreyman SJ, Michaels
JA, Thomas SM. Endovascular repair of abdominal aortic aneurysm.
Cochrane Database Syst Rev. 2014:CD004178.
26. Behrendt CA, Sedrakyan A, Rieß HC, Heidemann F, Kölbel T, Petersen
J, Debus ES. Short-term and long-term results of endovascular and
open repair of abdominal aortic aneurysms in Germany. J Vasc Surg.
2017;66:1704.e3–1711.e3. doi: 10.1016/j.jvs.2017.04.040
27. Baxter BT, Terrin MC, Dalman RL. Medical management of small ab-
dominal aortic aneurysms. Circulation. 2008;117:1883–1889. doi:
10.1161/CIRCULATIONAHA.107.735274
28. Ryer EJ, Garvin RP, Thomas B, Kuivaniemi H, Franklin DP, Elmore
JR. Patients with familial abdominal aortic aneurysms are at increased
risk for endoleak and secondary intervention following elective endo-
vascular aneurysm repair. J Vasc Surg. 2015;62:1119.e9–1124.e9. doi:
10.1016/j.jvs.2015.06.205
29. Lee R, Jones A, Cassimjee I, Handa A; Oxford Abdominal Aortic
Aneurysm Study. International opinion on priorities in research for
small abdominal aortic aneurysms and the potential path for research
to impact clinical management. Int J Cardiol. 2017;245:253–255. doi:
10.1016/j.ijcard.2017.06.058
30. Rizas KD, Ippagunta N, Tilson MD III. Immune cells and molecular me-
diators in the pathogenesis of the abdominal aortic aneurysm. Cardiol Rev.
2009;17:201–210. doi: 10.1097/CRD.0b013e3181b04698
31. Lysgaard Poulsen J, Stubbe J, Lindholt JS. Animal models used to explore
abdominal aortic aneurysms: a systematic review. Eur J Vasc Endovasc
Surg. 2016;52:487–499. doi: 10.1016/j.ejvs.2016.07.004
32. Sénémaud J, Caligiuri G, Etienne H, Delbosc S, Michel JB, Coscas R.
Translational relevance and recent advances of animal models of abdomi-
nal aortic aneurysm. Arterioscler Thromb Vasc Biol. 2017;37:401–410.
doi: 10.1161/ATVBAHA.116.308534
33. Azuma J, Asagami T, Dalman R, Tsao PS. Creation of murine experimen-
tal abdominal aortic aneurysms with elastase. J Vis Exp. 2009;29:1280.
34. White JV, Mazzacco SL. Formation and growth of aortic aneurysms in-
duced by adventitial elastolysis. Ann N Y Acad Sci. 1996;800:97–120.
35. Dwivedi AJ, Roy-Chaudhury P, Peden EK, Browne BJ, Ladenheim ED,
Scavo VA, Gustafson PN, Wong MD, Magill M, Lindow F, Blair AT, Jaff
MR, Franano FN, Burke SK. Application of human type I pancreatic e-
lastase (PRT-201) to the venous anastomosis of arteriovenous grafts in
patients with chronic kidney disease. J Vasc Access. 2014;15:376–384.
36. Bergamini TM, Seabrook GR, Bandyk DF, Towne JB. Symptomatic recur-
rent carotid stenosis and aneurysmal degeneration after endarterectomy.
Surgery. 1993;113:580–586.
37. Dobrin PB, Baker WH, Gley WC. Elastolytic and collagenolytic studies
of arteries. Implications for the mechanical properties of aneurysms. Arch
Surg. 1984;119:405–409.
38. Carsten CG III, Calton WC, Johanning JM, Armstrong PJ, Franklin DP,
Carey DJ, Elmore JR. Elastase is not sufficient to induce experimental
abdominal aortic aneurysms. J Vasc Surg. 2001;33:1255–1262.
39. Thompson RW, Curci JA, Ennis TL, Mao D, Pagano MB, Pham CT.
Pathophysiology of abdominal aortic aneurysms: insights from the elas-
tase-induced model in mice with different genetic backgrounds. Ann N Y
Acad Sci. 2006;1085:59–73. doi: 10.1196/annals.1383.029
40. Kokje VBC, Gäbel G, Koole D, Northoff BH, Holdt LM, Hamming
JF, Lindeman JHN. IL-6: a Janus-like factor in abdominal aor-
tic aneurysm disease. Atherosclerosis. 2016;251:139–146. doi:
10.1016/j.atherosclerosis.2016.06.021
41. Lareyre F, Clément M, Raffort J, Pohlod S, Patel M, Esposito B, Master
L, Finigan A, Vandestienne M, Stergiopulos N, Taleb S, Trachet B,
Mallat Z. TGFβ (Transforming Growth Factor-β) blockade induces a
Downloaded from http://ahajournals.org by on February 20, 2019
Lindeman and Matsumura Pharmacologic Management of Aneurysms 641
human-like disease in a nondissecting mouse model of abdominal aor-
tic aneurysm. Arterioscler Thromb Vasc Biol. 2017;37:2171–2181. doi:
10.1161/ATVBAHA.117.309999
42. Lu G, Su G, Davis JP, Schaheen B, Downs E, Roy RJ, Ailawadi G,
Upchurch GR Jr. A novel chronic advanced stage abdominal aor-
tic aneurysm murine model. J Vasc Surg. 2017;66:232.e4–242.e4. doi:
10.1016/j.jvs.2016.07.105
43. Yamanouchi D, Morgan S, Stair C, Seedial S, Lengfeld J, Kent KC,
Liu B. Accelerated aneurysmal dilation associated with apoptosis
and inflammation in a newly developed calcium phosphate rodent ab-
dominal aortic aneurysm model. J Vasc Surg. 2012;56:455–461. doi:
10.1016/j.jvs.2012.01.038
44. Saraff K, Babamusta F, Cassis LA, Daugherty A. Aortic dissection pre-
cedes formation of aneurysms and atherosclerosis in angiotensin II-
infused, apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol.
2003;23:1621–1626. doi: 10.1161/01.ATV.0000085631.76095.64
45. Trachet B, Aslanidou L, Piersigilli A, Fraga-Silva RA, Sordet-Dessimoz
J, Villanueva-Perez P, Stampanoni MFM, Stergiopulos N, Segers P.
Angiotensin II infusion into ApoE-/- mice: a model for aortic dissection
rather than abdominal aortic aneurysm? Cardiovasc Res. 2017;113:1230–
1242. doi: 10.1093/cvr/cvx128
46. Saito T, Hasegawa Y, Ishigaki Y, Yamada T, Gao J, Imai J, Uno K,
Kaneko K, Ogihara T, Shimosawa T, Asano T, Fujita T, Oka Y, Katagiri
H. Importance of endothelial NF-κB signalling in vascular remodelling
and aortic aneurysm formation. Cardiovasc Res. 2013;97:106–114. doi:
10.1093/cvr/cvs298
47. Yoshimura K, Aoki H, Ikeda Y, Fujii K, Akiyama N, Furutani A, Hoshii
Y, Tanaka N, Ricci R, Ishihara T, Esato K, Hamano K, Matsuzaki
M. Regression of abdominal aortic aneurysm by inhibition of c-Jun
N-terminal kinase. Nat Med. 2005;11:1330–1338. doi: 10.1038/nm1335
48. Wang YX, Martin-McNulty B, da Cunha V, et al. Fasudil, a Rho-
kinase inhibitor, attenuates angiotensin II-induced abdominal a-
ortic aneurysm in apolipoprotein E-deficient mice by inhibiting
apoptosis and proteolysis. Circulation. 2005;111:2219–2226. doi:
10.1161/01.CIR.0000163544.17221.BE
49. Johnston WF, Salmon M, Su G, Lu G, Stone ML, Zhao Y, Owens
GK, Upchurch GR Jr, Ailawadi G. Genetic and pharmacologic dis-
ruption of interleukin-1β signaling inhibits experimental aortic aneu-
rysm formation. Arterioscler Thromb Vasc Biol. 2013;33:294–304. doi:
10.1161/ATVBAHA.112.300432
50. Xiong W, MacTaggart J, Knispel R, Worth J, Persidsky Y, Baxter BT.
Blocking TNF-alpha attenuates aneurysm formation in a murine model. J
Immunol. 2009;183:2741–2746. doi: 10.4049/jimmunol.0803164
51. Wang Q, Ren J, Morgan S, Liu Z, Dou C, Liu B. Monocyte chemoattractant
protein-1 (MCP-1) regulates macrophage cytotoxicity in abdominal aortic
aneurysm. PLoS One. 2014;9:e92053. doi: 10.1371/journal.pone.0092053
52. Schaheen B, Downs EA, Serbulea V, Almenara CC, Spinosa M, Su G,
Zhao Y, Srikakulapu P, Butts C, McNamara CA, Leitinger N, Upchurch
GR Jr, Meher AK, Ailawadi G. B-cell depletion promotes aortic infiltra-
tion of immunosuppressive cells and is protective of experimental aor-
tic aneurysm. Arterioscler Thromb Vasc Biol. 2016;36:2191–2202. doi:
10.1161/ATVBAHA.116.307559
53. Zhang S, Kan X, Li Y, Li P, Zhang C, Li G, Du J, You B. Deficiency of
γδT cells protects against abdominal aortic aneurysms by regulating phos-
phoinositide 3-kinase/AKT signaling. J Vasc Surg. 2018;67:899.e1–908.
e1. doi: 10.1016/j.jvs.2016.03.474
54. Eliason JL, Hannawa KK, Ailawadi G, Sinha I, Ford JW, Deogracias MP,
Roelofs KJ, Woodrum DT, Ennis TL, Henke PK, Stanley JC, Thompson
RW, Upchurch GR Jr. Neutrophil depletion inhibits experimental abdom-
inal aortic aneurysm formation. Circulation. 2005;112:232–240. doi:
10.1161/CIRCULATIONAHA.104.517391
55. Inoue N, Muramatsu M, Jin D, Takai S, Hayashi T, Katayama H,
Kitaura Y, Tamai H, Miyazaki M. Effects of chymase inhibitor on an-
giotensin II-induced abdominal aortic aneurysm development in apo-
lipoprotein E-deficient mice. Atherosclerosis. 2009;204:359–364. doi:
10.1016/j.atherosclerosis.2008.09.032
56. Zhou HF, Yan H, Bertram P, Hu Y, Springer LE, Thompson RW, Curci
JA, Hourcade DE, Pham CT. Fibrinogen-specific antibody induces ab-
dominal aortic aneurysm in mice through complement lectin path-
way activation. Proc Natl Acad Sci USA. 2013;110:E4335–E4344. doi:
10.1073/pnas.1315512110
57. Zhou HF, Yan H, Stover CM, Fernandez TM, Rodriguez de Cordoba S,
Song WC, Wu X, Thompson RW, Schwaeble WJ, Atkinson JP, Hourcade
DE, Pham CT. Antibody directs properdin-dependent activation of
the complement alternative pathway in a mouse model of abdominal
aortic aneurysm. Proc Natl Acad Sci USA. 2012;109:E415–E422. doi:
10.1073/pnas.1119000109
58. Ahluwalia N, Lin AY, Tager AM, Pruitt IE, Anderson TJ, Kristo F, Shen D,
Cruz AR, Aikawa M, Luster AD, Gerszten RE. Inhibited aortic aneurysm
formation in BLT1-deficient mice. J Immunol. 2007;179:691–697.
59. Ghoshal S, Loftin CD. Cyclooxygenase-2 inhibition attenuates abdom-
inal aortic aneurysm progression in hyperlipidemic mice. PLoS One.
2012;7:e44369. doi: 10.1371/journal.pone.0044369
60. Marinkovic G, Hibender S, Hoogenboezem M, van Broekhoven A,
Girigorie AF, Bleeker N, Hamers AA, Stap J, van Buul JD, de Vries CJ,
de Waard V. Immunosuppressive drug azathioprine reduces aneurysm pro-
gression through inhibition of Rac1 and c-Jun-terminal-N-kinase in en-
dothelial cells. Arterioscler Thromb Vasc Biol. 2013;33:2380–2388. doi:
10.1161/ATVBAHA.113.301394
61. Yamaguchi T, Yokokawa M, Suzuki M, Higashide S, Katoh Y,
Sugiyama S, Misaki T. The effect of immunosuppression on aortic dil-
atation in a rat aneurysm model. Surg Today. 2000;30:1093–1099. doi:
10.1007/s005950070007
62. Ennis T, Jin J, Bartlett S, Arif B, Grapperhaus K, Curci JA. Effect
of novel limited-spectrum MMP inhibitor XL784 in abdominal aor-
tic aneurysms. J Cardiovasc Pharmacol Ther. 2012;17:417–426. doi:
10.1177/1074248412455695
63. Allaire E, Forough R, Clowes M, Starcher B, Clowes AW. Local overex-
pression of TIMP-1 prevents aortic aneurysm degeneration and rupture in
a rat model. J Clin Invest. 1998;102:1413–1420. doi: 10.1172/JCI2909
64. Qin Y, Cao X, Guo J, Zhang Y, Pan L, Zhang H, Li H, Tang C, Du J,
Shi GP. Deficiency of cathepsin S attenuates angiotensin II-induced ab-
dominal aortic aneurysm formation in apolipoprotein E-deficient mice.
Cardiovasc Res. 2012;96:401–410. doi: 10.1093/cvr/cvs263
65. Sun J, Sukhova GK, Zhang J, Chen H, Sjöberg S, Libby P, Xia M, Xiong
N, Gelb BD, Shi GP. Cathepsin K deficiency reduces elastase perfusion-
induced abdominal aortic aneurysms in mice. Arterioscler Thromb Vasc
Biol. 2012;32:15–23. doi: 10.1161/ATVBAHA.111.235002
66. Ang LS, Boivin WA, Williams SJ, Zhao H, Abraham T, Carmine-
Simmen K, McManus BM, Bleackley RC, Granville DJ. Serpina3n
attenuates granzyme B-mediated decorin cleavage and rupture in a
murine model of aortic aneurysm. Cell Death Dis. 2011;2:e209. doi:
10.1038/cddis.2011.88
67. Deng GG, Martin-McNulty B, Sukovich DA, Freay A, Halks-Miller M,
Thinnes T, Loskutoff DJ, Carmeliet P, Dole WP, Wang YX. Urokinase-
type plasminogen activator plays a critical role in angiotensin II-
induced abdominal aortic aneurysm. Circ Res. 2003;92:510–517. doi:
10.1161/01.RES.0000061571.49375.E1
68. Yu Z, Morimoto K, Yu J, Bao W, Okita Y, Okada K. Endogenous superox-
ide dismutase activation by oral administration of riboflavin reduces ab-
dominal aortic aneurysm formation in rats. J Vasc Surg. 2016;64:737–745.
doi: 10.1016/j.jvs.2015.03.045
69. Maiellaro-Rafferty K, Weiss D, Joseph G, Wan W, Gleason RL, Taylor
WR. Catalase overexpression in aortic smooth muscle prevents patho-
logical mechanical changes underlying abdominal aortic aneurysm for-
mation. Am J Physiol Heart Circ Physiol. 2011;301:H355–H362. doi:
10.1152/ajpheart.00040.2011
70. Morimoto K, Hasegawa T, Tanaka A, Wulan B, Yu J, Morimoto N, Okita
Y, Okada K. Free-radical scavenger edaravone inhibits both formation
and development of abdominal aortic aneurysm in rats. J Vasc Surg.
2012;55:1749–1758. doi: 10.1016/j.jvs.2011.11.059
71. Gavrila D, Li WG, McCormick ML, Thomas M, Daugherty A, Cassis
LA, Miller FJ Jr, Oberley LW, Dellsperger KC, Weintraub NL. Vitamin
E inhibits abdominal aortic aneurysm formation in angiotensin II-
infused apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol.
2005;25:1671–1677. doi: 10.1161/01.ATV.0000172631.50972.0f
72. Slaiby JM, Ricci MA, Gadowski GR, Hendley ED, Pilcher DB. Expansion
of aortic aneurysms is reduced by propranolol in a hypertensive rat model.
J Vasc Surg. 1994;20:178–183.
73. Miao XN, Siu KL, Cai H. Nifedipine attenuation of abdominal a-
ortic aneurysm in hypertensive and non-hypertensive mice: mecha-
nisms and implications. J Mol Cell Cardiol. 2015;87:152–159. doi:
10.1016/j.yjmcc.2015.07.031
74. Liao S, Miralles M, Kelley BJ, Curci JA, Borhani M, Thompson RW.
Suppression of experimental abdominal aortic aneurysms in the rat by
treatment with angiotensin-converting enzyme inhibitors. J Vasc Surg.
2001;33:1057–1064. doi: 10.1067/mva.2001.112810
75. Xiong F, Zhao J, Zeng G, Huang B, Yuan D, Yang Y. Inhibition of AAA in a
rat model by treatment with ACEI perindopril. J Surg Res. 2014;189:166–
173. doi: 10.1016/j.jss.2014.01.057
Downloaded from http://ahajournals.org by on February 20, 2019
642 Circulation Research February 15, 2019
76. Iida Y, Xu B, Schultz GM, Chow V, White JJ, Sulaimon S, Hezi-Yamit
A, Peterson SR, Dalman RL. Efficacy and mechanism of angiotensin II
receptor blocker treatment in experimental abdominal aortic aneurysms.
PLoS One. 2012;7:e49642. doi: 10.1371/journal.pone.0049642
77. Lizarbe TR, Tarín C, Gómez M, Lavin B, Aracil E, Orte LM, Zaragoza
C. Nitric oxide induces the progression of abdominal aortic aneurysms
through the matrix metalloproteinase inducer EMMPRIN. Am J Pathol.
2009;175:1421–1430. doi: 10.2353/ajpath.2009.080845
78. Kalyanasundaram A, Elmore JR, Manazer JR, Golden A, Franklin DP,
Galt SW, Zakhary EM, Carey DJ. Simvastatin suppresses experimen-
tal aortic aneurysm expansion. J Vasc Surg. 2006;43:117–124. doi:
10.1016/j.jvs.2005.08.007
79. Shiraya S, Miyake T, Aoki M, Yoshikazu F, Ohgi S, Nishimura M,
Ogihara T, Morishita R. Inhibition of development of experimental a-
ortic abdominal aneurysm in rat model by atorvastatin through inhibi-
tion of macrophage migration. Atherosclerosis. 2009;202:34–40. doi:
10.1016/j.atherosclerosis.2008.03.020
80. Delbosc S, Rouer M, Alsac JM, Louedec L, Al Shoukr F, Rouzet F, Michel
JB, Meilhac O. High-density lipoprotein therapy inhibits Porphyromonas
gingivalis-induced abdominal aortic aneurysm progression. Thromb
Haemost. 2016;115:789–799. doi: 10.1160/TH15-05-0398
81. Escudero P, Navarro A, Ferrando C, Furio E, Gonzalez-Navarro H, Juez
M, Sanz MJ, Piqueras L. Combined treatment with bexarotene and ro-
suvastatin reduces angiotensin-II-induced abdominal aortic aneurysm in
apoE(-/-) mice and angiogenesis. Br J Pharmacol. 2015;172:2946–2960.
doi: 10.1111/bph.13098
82. Golledge J, Cullen B, Rush C, Moran CS, Secomb E, Wood F, Daugherty
A, Campbell JH, Norman PE. Peroxisome proliferator-activated recep-
tor ligands reduce aortic dilatation in a mouse model of aortic aneurysm.
Atherosclerosis. 2010;210:51–56. doi: 10.1016/j.atherosclerosis.2009.10.027
83. Yamawaki-Ogata A, Fu X, Hashizume R, Fujimoto KL, Araki Y, Oshima
H, Narita Y, Usui A. Therapeutic potential of bone marrow-derived mes-
enchymal stem cells in formed aortic aneurysms of a mouse model. Eur J
Cardiothorac Surg. 2014;45:e156–e165. doi: 10.1093/ejcts/ezu018
84. Blose KJ, Ennis TL, Arif B, Weinbaum JS, Curci JA, Vorp DA.
Periadventitial adipose-derived stem cell treatment halts elastase-induced
abdominal aortic aneurysm progression. Regen Med. 2014;9:733–741.
doi: 10.2217/rme.14.61
85. Giraud A, Zeboudj L, Vandestienne M, Joffre J, Esposito B, Potteaux S,
Vilar J, Cabuzu D, Kluwe J, Seguier S, Tedgui A, Mallat Z, Lafont A,
Ait-Oufella H. Gingival fibroblasts protect against experimental abdom-
inal aortic aneurysm development and rupture through tissue inhibitor of
metalloproteinase-1 production. Cardiovasc Res. 2017;113:1364–1375.
doi: 10.1093/cvr/cvx110
86. Gao F, Chambon P, Offermanns S, Tellides G, Kong W, Zhang X, Li W.
Disruption of TGF-β signaling in smooth muscle cell prevents elastase-
induced abdominal aortic aneurysm. Biochem Biophys Res Commun.
2014;454:137–143. doi: 10.1016/j.bbrc.2014.10.053
87. Cheng J, Koenig SN, Kuivaniemi HS, Garg V, Hans CP. Pharmacological
inhibitor of notch signaling stabilizes the progression of small abdominal
aortic aneurysm in a mouse model. J Am Heart Assoc. 2014;3:e001064.
doi: 10.1161/JAHA.114.001064
88. Krishna SM, Seto SW, Jose RJ, Li J, Morton SK, Biros E, Wang Y,
Nsengiyumva V, Lindeman JH, Loots GG, Rush CM, Craig JM, Golledge
J. Wnt signaling pathway inhibitor sclerostin inhibits angiotensin II-
induced aortic aneurysm and atherosclerosis. Arterioscler Thromb Vasc
Biol. 2017;37:553–566. doi: 10.1161/ATVBAHA.116.308723
89. Krishna SM, Seto SW, Jose RJ, Biros E, Moran CS, Wang Y, Clancy P,
Golledge J. A peptide antagonist of thrombospondin-1 promotes abdomi-
nal aortic aneurysm progression in the angiotensin II-infused apolipopro-
tein-E-deficient mouse. Arterioscler Thromb Vasc Biol. 2015;35:389–398.
doi: 10.1161/ATVBAHA.114.304732
90. Obama T, Tsuji T, Kobayashi T, Fukuda Y, Takayanagi T, Taro Y, Kawai
T, Forrester SJ, Elliott KJ, Choi E, Daugherty A, Rizzo V, Eguchi S.
Epidermal growth factor receptor inhibitor protects against abdominal a-
ortic aneurysm in a mouse model. Clin Sci (Lond). 2015;128:559–565.
doi: 10.1042/CS20140696
91. Yang L, Shen L, Li G, Yuan H, Jin X, Wu X. Silencing of hypoxia in-
ducible factor-1α gene attenuated angiotensin -induced abdominal
aortic aneurysm in apolipoprotein E-deficient mice. Atherosclerosis.
2016;252:40–49. doi: 10.1016/j.atherosclerosis.2016.07.010
92. Yang L, Shen L, Gao P, Li G, He Y, Wang M, Zhou H, Yuan H, Jin
X, Wu X. Effect of AMPK signal pathway on pathogenesis of ab-
dominal aortic aneurysms. Oncotarget. 2017;8:92827–92840. doi:
10.18632/oncotarget.21608
93. Wang C, Wang Y, Yu M, Chen C, Xu L, Cao Y, Qi R. Grape-seed poly-
phenols play a protective role in elastase-induced abdominal aortic aneu-
rysm in mice. Sci Rep. 2017;7:9402. doi: 10.1038/s41598-017-09674-4
94. Lu G, Su G, Zhao Y, Johnston WF, Sherman NE, Rissman EF, Lau C,
Ailawadi G, Upchurch GR Jr. Dietary phytoestrogens inhibit experi-
mental aneurysm formation in male mice. J Surg Res. 2014;188:326–
338. doi: 10.1016/j.jss.2013.11.1108
95. Zhang X, Thatcher S, Wu C, Daugherty A, Cassis LA. Castration of
male mice prevents the progression of established angiotensin II-induced
abdominal aortic aneurysms. J Vasc Surg. 2015;61:767–776. doi:
10.1016/j.jvs.2013.11.004
96. Martin-McNulty B, Tham DM, da Cunha V, Ho JJ, Wilson DW, Rutledge
JC, Deng GG, Vergona R, Sullivan ME, Wang YX. 17 Beta-estradiol
attenuates development of angiotensin II-induced aortic abdominal aneu-
rysm in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol.
2003;23:1627–1632. doi: 10.1161/01.ATV.0000085842.20866.6A
97. Liu S, Xie Z, Daugherty A, Cassis LA, Pearson KJ, Gong MC, Guo Z.
Mineralocorticoid receptor agonists induce mouse aortic aneurysm for-
mation and rupture in the presence of high salt. Arterioscler Thromb Vasc
Biol. 2013;33:1568–1579. doi: 10.1161/ATVBAHA.112.300820
98. Cao RY, St Amand T, Li X, Yoon SH, Wang CP, Song H, Maruyama
T, Brown PM, Zelt DT, Funk CD. Prostaglandin receptor EP4 in ab-
dominal aortic aneurysms. Am J Pathol. 2012;181:313–321. doi:
10.1016/j.ajpath.2012.03.016
99. Xie X, Lu H, Moorleghen JJ, Howatt DA, Rateri DL, Cassis LA,
Daugherty A. Doxycycline does not influence established abdom-
inal aortic aneurysms in angiotensin II-infused mice. PLoS One.
2012;7:e46411. doi: 10.1371/journal.pone.0046411
100. Golledge J, Cullen B, Moran C, Rush C. Efficacy of simvas-
tatin in reducing aortic dilatation in mouse models of abdominal
aortic aneurysm. Cardiovasc Drugs Ther. 2010;24:373–378. doi:
10.1007/s10557-010-6262-8
101. Bai L, Beckers L, Wijnands E, Lutgens SP, Herías MV, Saftig P, Daemen
MJ, Cleutjens K, Lutgens E, Biessen EA, Heeneman S. Cathepsin K gene
disruption does not affect murine aneurysm formation. Atherosclerosis.
2010;209:96–103. doi: 10.1016/j.atherosclerosis.2009.09.001
102. Hingorani A, Ascher E, Scheinman M, Yorkovich W, DePippo P, Ladoulis
CT, Salles-Cunha S. The effect of tumor necrosis factor binding protein
and interleukin-1 receptor antagonist on the development of abdominal
aortic aneurysms in a rat model. J Vasc Surg. 1998;28:522–526.
103. Easterbrook PJ, Berlin JA, Gopalan R, Matthews DR. Publication bias in
clinical research. Lancet. 1991;337:867–872.
104. Trachet B, Fraga-Silva RA, Jacquet PA, Stergiopulos N, Segers P.
Incidence, severity, mortality, and confounding factors for dissect-
ing AAA detection in angiotensin II-infused mice: a meta-analysis.
Cardiovasc Res. 2015;108:159–170. doi: 10.1093/cvr/cvv215
105. Freiberg MS, Arnold AM, Newman AB, Edwards MS, Kraemer
KL, Kuller LH. Abdominal aortic aneurysms, increasing infra-
renal aortic diameter, and risk of total mortality and incident car-
diovascular disease events: 10-year follow-up data from the
Cardiovascular Health Study. Circulation. 2008;117:1010–1017. doi:
10.1161/CIRCULATIONAHA.107.720219
106. Forsdahl SH, Solberg S, Singh K, Jacobsen BK. Abdominal aortic aneu-
rysms, or a relatively large diameter of non-aneurysmal aortas, increase
total and cardiovascular mortality: the Tromsø study. Int J Epidemiol.
2010;39:225–232. doi: 10.1093/ije/dyp320
107. Karthikesalingam A, Bahia SS, Patterson BO, Peach G, Vidal-Diez A,
Ray KK, Sharma R, Hinchliffe RJ, Holt PJ, Thompson MM. The short-
fall in long-term survival of patients with repaired thoracic or abdom-
inal aortic aneurysms: retrospective case-control analysis of hospital
episode statistics. Eur J Vasc Endovasc Surg. 2013;46:533–541. doi:
10.1016/j.ejvs.2013.09.008
108. Baumgartner I, Hirsch AT, Abola MT, Cacoub PP, Poldermans D,
Steg PG, Creager MA, Bhatt DL; REACH Registry Investigators.
Cardiovascular risk profile and outcome of patients with abdominal aortic
aneurysm in out-patients with atherothrombosis: data from the Reduction
of Atherothrombosis for Continued Health (REACH) Registry. J Vasc
Surg. 2008;48:808–814. doi: 10.1016/j.jvs.2008.05.026
109. Welten GM, Schouten O, Hoeks SE, Chonchol M, Vidakovic R, van
Domburg RT, Bax JJ, van Sambeek MR, Poldermans D. Long-term prog-
nosis of patients with peripheral arterial disease: a comparison in patients
with coronary artery disease. J Am Coll Cardiol. 2008;51:1588–1596.
doi: 10.1016/j.jacc.2007.11.077
110. Parkinson F, Ferguson S, Lewis P, Williams IM, Twine CP; South
East Wales Vascular Network. Rupture rates of untreated large
Downloaded from http://ahajournals.org by on February 20, 2019
Lindeman and Matsumura Pharmacologic Management of Aneurysms 643
abdominal aortic aneurysms in patients unfit for elective repair. J Vasc
Surg. 2015;61:1606–1612. doi: 10.1016/j.jvs.2014.10.023
111. Bulder RMA, Bastiaannet E, Hamming JF, Lindeman JH. Equal long-
term survival after elective endovascular or open AAA repair: a system-
atic review and meta-analysis. Br J Surg. In press.
112. Huang Q, Yang H, Lin Q, Hu M, Meng Y, Qin X. Effect of statin
therapy on survival after abdominal aortic aneurysm repair: a system-
atic review and meta-analysis. World J Surg. 2018;42:3443–3450. doi:
10.1007/s00268-018-4586-x
113. Mathisen SR, Abdelnoor M. Beneficial effect of statins on total mortality
in abdominal aortic aneurysm (AAA) repair. Vasc Med. 2017;22:406–
410. doi: 10.1177/1358863X17724221
114. Leach SD, Toole AL, Stern H, DeNatale RW, Tilson MD. Effect of be-
ta-adrenergic blockade on the growth rate of abdominal aortic aneu-
rysms. Arch Surg. 1988;123:606–609.
115. Gadowski GR, Pilcher DB, Ricci MA. Abdominal aortic aneurysm ex-
pansion rate: effect of size and beta-adrenergic blockade. J Vasc Surg.
1994;19:727–731.
116. Wilmink AB, Vardulaki KA, Hubbard CS, Day NE, Ashton HA, Scott
AP, Quick CR. Are antihypertensive drugs associated with abdominal
aortic aneurysms? J Vasc Surg. 2002;36:751–757.
117. Bhak RH, Wininger M, Johnson GR, Lederle FA, Messina LM, Ballard
DJ, Wilson SE; Aneurysm Detection and Management (ADAM) Study
Group. Factors associated with small abdominal aortic aneurysm expan-
sion rate. JAMA Surg. 2015;150:44–50. doi: 10.1001/jamasurg.2014.2025
118. Lindholt JS, Henneberg EW, Juul S, Fasting H. Impaired results of a
randomised double blinded clinical trial of propranolol versus placebo
on the expansion rate of small abdominal aortic aneurysms. Int Angiol.
1999;18:52–57.
119. Propranolol Aneurysm Trial Investigators. Propranolol for small ab-
dominal aortic aneurysms: results of a randomized trial. J Vasc Surg.
2002;35:72–79. doi: 10.1067/mva.2002.121308
120. Hackam DG, Thiruchelvam D, Redelmeier DA. Angiotensin-converting
enzyme inhibitors and aortic rupture: a population-based case-control
study. Lancet. 2006;368:659–665. doi: 10.1016/S0140-6736(06)69250-7
121. Kortekaas KE, Meijer CA, Hinnen JW, Dalman RL, Xu B, Hamming
JF, Lindeman JH. ACE inhibitors potently reduce vascular inflammation,
results of an open proof-of-concept study in the abdominal aortic aneu-
rysm. PLoS One. 2014;9:e111952. doi: 10.1371/journal.pone.0111952
122. Thompson AR, Cooper JA, Ashton HA, Hafez H. Growth rates of small
abdominal aortic aneurysms correlate with clinical events. Br J Surg.
2010;97:37–44. doi: 10.1002/bjs.6779
123. Sweeting MJ, Thompson SG, Brown LC, Greenhalgh RM, Powell JT.
Use of angiotensin converting enzyme inhibitors is associated with
increased growth rate of abdominal aortic aneurysms. J Vasc Surg.
2010;52:1–4. doi: 10.1016/j.jvs.2010.02.264
124. Bicknell CD, Kiru G, Falaschetti E, Powell JT, Poulter NR; AARDVARK
Collaborators. An evaluation of the effect of an angiotensin-converting
enzyme inhibitor on the growth rate of small abdominal aortic aneu-
rysms: a randomized placebo-controlled trial (AARDVARK). Eur Heart
J. 2016;37:3213–3221. doi: 10.1093/eurheartj/ehw257
125. Morris DR, Cunningham MA, Ahimastos AA, et al. Telmisartan in
the Management of Abdominal Aortic Aneurysm (TEDY): the study
protocol for a randomized controlled trial. Trials. 2015;16:274. doi:
10.1186/s13063-015-0793-z
126. Rompe F, Artuc M, Hallberg A, et al. Direct angiotensin II type 2 receptor
stimulation acts anti-inflammatory through epoxyeicosatrienoic acid and
inhibition of nuclear factor kappaB. Hypertension. 2010;55:924–931.
doi: 10.1161/HYPERTENSIONAHA.109.147843
127. Rehman A, Leibowitz A, Yamamoto N, Rautureau Y, Paradis P,
Schiffrin EL. Angiotensin type 2 receptor agonist compound 21
reduces vascular injury and myocardial fibrosis in stroke-prone spon-
taneously hypertensive rats. Hypertension. 2012;59:291–299. doi:
10.1161/HYPERTENSIONAHA.111.180158
128. Thompson A, Cooper JA, Fabricius M, Humphries SE, Ashton HA,
Hafez H. An analysis of drug modulation of abdominal aortic aneurysm
growth through 25 years of surveillance. J Vasc Surg. 2010;52:55.e2–61.
e2. doi: 10.1016/j.jvs.2010.02.012
129. Sukhija R, Aronow WS, Sandhu R, Kakar P, Babu S. Mortality and size
of abdominal aortic aneurysm at long-term follow-up of patients not
treated surgically and treated with and without statins. Am J Cardiol.
2006;97:279–280. doi: 10.1016/j.amjcard.2005.08.033
130. Schouten O, van Laanen JH, Boersma E, Vidakovic R, Feringa
HH, Dunkelgrün M, Bax JJ, Koning J, van Urk H, Poldermans D.
Statins are associated with a reduced infrarenal abdominal aortic
aneurysm growth. Eur J Vasc Endovasc Surg. 2006;32:21–26. doi:
10.1016/j.ejvs.2005.12.024
131. Schlösser FJ, Tangelder MJ, Verhagen HJ, van der Heijden GJ, Muhs
BE, van der Graaf Y, Moll FL; SMART study group. Growth predic-
tors and prognosis of small abdominal aortic aneurysms. J Vasc Surg.
2008;47:1127–1133. doi: 10.1016/j.jvs.2008.01.041
132. Karlsson L, Bergqvist D, Lindbäck J, Pärsson H. Expansion of small-
diameter abdominal aortic aneurysms is not reflected by the release of
inflammatory mediators IL-6, MMP-9 and CRP in plasma. Eur J Vasc
Endovasc Surg. 2009;37:420–424. doi: 10.1016/j.ejvs.2008.11.027
133. Karrowni W, Dughman S, Hajj GP, Miller FJ Jr. Statin therapy reduces
growth of abdominal aortic aneurysms. J Investig Med. 2011;59:1239–
1243. doi: 10.2130/JIM.0b013e31823548e8
134. Periard D, Guessous I, Mazzolai L, Haesler E, Monney P, Hayoz D.
Reduction of small infrarenal abdominal aortic aneurysm expansion rate
by statins. Vasa. 2012;41:35–42. doi: 10.1024/0301-1526/a000161
135. Mosorin M, Niemelä E, Heikkinen J, Lahtinen J, Tiozzo V, Satta J,
Juvonen T, Biancari F. The use of statins and fate of small abdominal
aortic aneurysms. Interact Cardiovasc Thorac Surg. 2008;7:578–581.
doi: 10.1510/icvts.2008.178103
136. Ferguson CD, Clancy P, Bourke B, Walker PJ, Dear A, Buckenham T,
Norman P, Golledge J. Association of statin prescription with small ab-
dominal aortic aneurysm progression. Am Heart J. 2010;159:307–313.
doi: 10.1016/j.ahj.2009.11.016
137. van der Meij E, Koning GG, Vriens PW, Peeters MF, Meijer CA,
Kortekaas KE, Dalman RL, van Bockel JH, Hanemaaijer R, Kooistra
T, Kleemann R, Lindeman JH. A clinical evaluation of statin pleiotropy:
statins selectively and dose-dependently reduce vascular inflammation.
PLoS One. 2013;8:e53882. doi: 10.1371/journal.pone.0053882
138. Lindholt JS, Sorensen HT, Michel JB, Thomsen HF, Henneberg EW.
Low-dose aspirin may prevent growth and later surgical repair of
medium-sized abdominal aortic aneurysms. Vasc Endovascular Surg.
2008;42:329–334. doi: 10.1177/1538574408315205
139. Bown MJ, Sweeting MJ, Brown LC, Powell JT, Thompson SG;
RESCAN Collaborators. Surveillance intervals for small abdominal
aortic aneurysms: a meta-analysis. JAMA. 2013;309:806–813. doi:
10.1001/jama.2013.950
140. Karlsson L, Gnarpe J, Bergqvist D, Lindbäck J, Pärsson H. The
effect of azithromycin and Chlamydophilia pneumonia infection
on expansion of small abdominal aortic aneurysms–a prospective
randomized double-blind trial. J Vasc Surg. 2009;50:23–29. doi:
10.1016/j.jvs.2008.12.048
141. Franklin IJ, Walton LJ, Brown L, Greenhalgh RN, Powell JT. Vascular
surgical society of great britain and ireland: non-steroidal anti-inflamma-
tory drugs to treat abdominal aortic aneurysm. Br J Surg. 1999;86:707.
doi: 10.1046/j.1365-2168.1999.0707b.x
142. Patel K, Zafar MA, Ziganshin BA, Elefteriades JA. Diabetes melli-
tus: is it protective against aneurysm? A narrative review. Cardiology.
2018;141:107–122. doi: 10.1159/000490373
143. Fujimura N, Xiong J, Kettler EB, Xuan H, Glover KJ, Mell MW, Xu
B, Dalman RL. Metformin treatment status and abdominal aortic an-
eurysm disease progression. J Vasc Surg. 2016;64:46.e8–54.e8. doi:
10.1016/j.jvs.2016.02.020
144. Itoga NK, Rothenberg KA, Suarez P, Ho TV, Mell MW, Xu B,
Curtin CM, Dalman RL. Metformin prescription status and abdom-
inal aortic aneurysm disease progression in the U.S. veteran pop-
ulation [published online September 6, 2018]. J Vasc Surg. doi:
10.1016/j.jvs.2018.06.194
145. Golledge J, Moxon J, Pinchbeck J, Anderson G, Rowbotham S, Jenkins
J, Bourke M, Bourke B, Dear A, Buckenham T, Jones R, Norman PE.
Association between metformin prescription and growth rates of ab-
dominal aortic aneurysms. Br J Surg. 2017;104:1486–1493. doi:
10.1002/bjs.10587
146. Vammen S, Lindholt JS, Ostergaard L, Fasting H, Henneberg EW.
Randomized double-blind controlled trial of roxithromycin for preven-
tion of abdominal aortic aneurysm expansion. Br J Surg. 2001;88:1066–
1072. doi: 10.1046/j.0007-1323.2001.01845.x
147. Høgh A, Vammen S, Ostergaard L, Joensen JB, Henneberg EW, Lindholt
JS. Intermittent roxithromycin for preventing progression of small ab-
dominal aortic aneurysms: long-term results of a small clinical trial. Vasc
Endovascular Surg. 2009;43:452–456. doi: 10.1177/1538574409335037
148. Hanemaaijer R, Visser H, Koolwijk P, Sorsa T, Salo T, Golub LM, van
Hinsbergh VW. Inhibition of MMP synthesis by doxycycline and chem-
ically modified tetracyclines (CMTs) in human endothelial cells. Adv
Dent Res. 1998;12:114–118. doi: 10.1177/08959374980120010301
Downloaded from http://ahajournals.org by on February 20, 2019
644 Circulation Research February 15, 2019
149. Golub LM, Sorsa T, Lee HM, Ciancio S, Sorbi D, Ramamurthy NS,
Gruber B, Salo T, Konttinen YT. Doxycycline inhibits neutrophil (PMN)-
type matrix metalloproteinases in human adult periodontitis gingiva. J
Clin Periodontol. 1995;22:100–109.
150. Curci JA, Petrinec D, Liao S, Golub LM, Thompson RW. Pharmacologic
suppression of experimental abdominal aortic aneurysms: acomparison
of doxycycline and four chemically modified tetracyclines. J Vasc Surg.
1998;28:1082–1093.
151. Yu M, Chen C, Cao Y, Qi R. Inhibitory effects of doxycycline on the onset
and progression of abdominal aortic aneurysm and its related mechanisms.
Eur J Pharmacol. 2017;811:101–109. doi: 10.1016/j.ejphar.2017.05.041
152. Curci JA, Mao D, Bohner DG, Allen BT, Rubin BG, Reilly JM, Sicard
GA, Thompson RW. Preoperative treatment with doxycycline reduces
aortic wall expression and activation of matrix metalloproteinases in
patients with abdominal aortic aneurysms. J Vasc Surg. 2000;31:325–342.
153. Lindeman JH. The pathophysiologic basis of abdominal aortic aneu-
rysm progression: a critical appraisal. Expert Rev Cardiovasc Ther.
2015;13:839–851. doi: 10.1586/14779072.2015.1052408
154. Mosorin M, Juvonen J, Biancari F, Satta J, Surcel HM, Leinonen M,
Saikku P, Juvonen T. Use of doxycycline to decrease the growth rate of ab-
dominal aortic aneurysms: a randomized, double-blind, placebo-controlled
pilot study. J Vasc Surg. 2001;34:606–610. doi: 10.1067/mva.2001.117891
155. Baxter BT, Pearce WH, Waltke EA, Littooy FN, Hallett JW Jr, Kent KC,
Upchurch GR Jr, Chaikof EL, Mills JL, Fleckten B, Longo GM, Lee JK,
Thompson RW. Prolonged administration of doxycycline in patients with
small asymptomatic abdominal aortic aneurysms: report of a prospective
(Phase II) multicenter study. J Vasc Surg. 2002;36:1–12.
156. Meijer CA, Stijnen T, Wasser MN, Hamming JF, van Bockel JH,
Lindeman JH; Pharmaceutical Aneurysm Stabilisation Trial Study
Group. Doxycycline for stabilization of abdominal aortic aneu-
rysms: a randomized trial. Ann Intern Med. 2013;159:815–823. doi:
10.7326/0003-4819-159-12-201312170-00007
157. Terrin M. Non-Invasive Treatment of Abdominal Aortic Aneurysm
Clinical Trial (N-TA^3CT). https://clinicaltrials.gov/ct2/show/
NCT01756833. Accessed January 30, 2019.
158. Abdul-Hussien H, Hanemaaijer R, Verheijen JH, van Bockel JH,
Geelkerken RH, Lindeman JH. Doxycycline therapy for abdominal an-
eurysm: improved proteolytic balance through reduced neutrophil con-
tent. J Vasc Surg. 2009;49:741–749. doi: 10.1016/j.jvs.2008.09.055
159. Lindeman JH, Abdul-Hussien H, van Bockel JH, Wolterbeek R,
Kleemann R. Clinical trial of doxycycline for matrix metalloproteinase-9
inhibition in patients with an abdominal aneurysm: doxycycline selec-
tively depletes aortic wall neutrophils and cytotoxic T cells. Circulation.
2009;119:2209–2216. doi: 10.1161/CIRCULATIONAHA.108.806505
160. Fujimiya H, Nakashima S, Miyata H, Nozawa Y. Effect of a novel antial-
lergic drug, pemirolast, on activation of rat peritoneal mast cells: inhi-
bition of exocytotic response and membrane phospholipid turnover. Int
Arch Allergy Appl Immunol. 1991;96:62–67.
161. Sillesen H, Eldrup N, Hultgren R, Lindeman J, Bredahl K, Thompson
M, Wanhainen A, Wingren U, Swedenborg J; AORTA Trial Investigators.
Randomized clinical trial of mast cell inhibition in patients with a
medium-sized abdominal aortic aneurysm. Br J Surg. 2015;102:894–
901. doi: 10.1002/bjs.9824
162. Novartis. ACZ885 for the Treatment of Abdominal Aortic Aneurysm
(AAA). https://clinicaltrials.gov/ct2/show/results/NCT02007252. Accessed
January 30, 2019.
163. Motoki T, Kurobe H, Hirata Y, Nakayama T, Kinoshita H, Rocco KA,
Sogabe H, Hori T, Sata M, Kitagawa T. PPAR-γ agonist attenuates in-
flammation in aortic aneurysm patients. Gen Thorac Cardiovasc Surg.
2015;63:565–571. doi: 10.1007/s11748-015-0576-1
164. Pinchbeck JL, Moxon JV, Rowbotham SE, et al. Randomized placebo-con-
trolled trial assessing the effect of 24-week fenofibrate therapy on circulat-
ing markers of abdominal aortic aneurysm: outcomes from the FAME -2
trial. J Am Heart Assoc. 2018;7:e009866. doi: 10.1161/JAHA.118.009866
165. Nieuwland AJ, Kokje VB, Koning OH, Hamming JF, Szuhai K, Claas
FH, Lindeman JH. Activation of the vitamin D receptor selectively
interferes with calcineurin-mediated inflammation: a clinical evaluation
in the abdominal aortic aneurysm. Lab Invest. 2016;96:784–790. doi:
10.1038/labinvest.2016.55
166. Deeg MA, Meijer CA, Chan LS, Shen L, Lindeman JH. Prognostic and
predictive biomarkers of abdominal aortic aneurysm growth rate. Curr
Med Res Opin. 2016;32:509–517. doi: 10.1185/03007995.2015.1128406
167. Dawson JA, Choke E, Loftus IM, Cockerill GW, Thompson MM.
A randomised placebo-controlled double-blind trial to evaluate
lipid-lowering pharmacotherapy on proteolysis and inflammation in
abdominal aortic aneurysms. Eur J Vasc Endovasc Surg. 2011;41:28–35.
doi: 10.1016/j.ejvs.2010.08.023
168. Franklin IJ, Walton LJ, Greenhalgh RM, Powell JT. The influence of
indomethacin on the metabolism and cytokine secretion of human
aneurysmal aorta. Eur J Vasc Endovasc Surg. 1999;18:35–42. doi:
10.1053/ejvs.1999.0820
169. Abisi S, Burnand KG, Humphries J, Waltham M, Taylor P, Smith A.
Effect of statins on proteolytic activity in the wall of abdominal aortic
aneurysms. Br J Surg. 2008;95:333–337. doi: 10.1002/bjs.5989
170. Wilson WR, Evans J, Bell PR, Thompson MM. HMG-CoA reductase
inhibitors (statins) decrease MMP-3 and MMP-9 concentrations in ab-
dominal aortic aneurysms. Eur J Vasc Endovasc Surg. 2005;30:259–262.
doi: 10.1016/j.ejvs.2005.02.044
171. Evans J, Powell JT, Schwalbe E, Loftus IM, Thompson MM. Simvastatin
attenuates the activity of matrix metalloprotease-9 in aneurysmal
aortic tissue. Eur J Vasc Endovasc Surg. 2007;34:302–303. doi:
10.1016/j.ejvs.2007.04.011
172. Hurks R, Hoefer IE, Vink A, Pasterkamp G, Schoneveld A, Kerver M,
de Vries JP, Tangelder MJ, Moll FL. Different effects of commonly pre-
scribed statins on abdominal aortic aneurysm wall biology. Eur J Vasc
Endovasc Surg. 2010;39:569–576. doi: 10.1016/j.ejvs.2010.01.030
173. Schweitzer M, Mitmaker B, Obrand D, Sheiner N, Abraham C, Dostanic
S, Meilleur M, Sugahara T, Chalifour LE. Atorvastatin modulates ma-
trix metalloproteinase expression, activity, and signaling in abdominal
aortic aneurysms. Vasc Endovascular Surg. 2010;44:116–122. doi:
10.1177/1538574409348352
174. Kajimoto K, Miyauchi K, Kasai T, Shimada K, Kojima Y, Shimada A,
Niinami H, Amano A, Daida H. Short-term 20-mg atorvastatin therapy
reduces key inflammatory factors including c-Jun N-terminal kinase
and dendritic cells and matrix metalloproteinase expression in human
abdominal aortic aneurysmal wall. Atherosclerosis. 2009;206:505–511.
doi: 10.1016/j.atherosclerosis.2009.03.028
175. Piechota-Polanczyk A, Goraca A, Demyanets S, Mittlboeck M, Domenig
C, Neumayer C, Wojta J, Nanobachvili J, Huk I, Klinger M. Simvastatin
decreases free radicals formation in the human abdominal aortic aneu-
rysm wall via NF-κB. Eur J Vasc Endovasc Surg. 2012;44:133–137. doi:
10.1016/j.ejvs.2012.04.020
176. Yoshimura K, Nagasawa A, Kudo J, Onoda M, Morikage N, Furutani A,
Aoki H, Hamano K. Inhibitory effect of statins on inflammation-related
pathways in human abdominal aortic aneurysm tissue. Int J Mol Sci.
2015;16:11213–11228. doi: 10.3390/ijms160511213
177. Lindeman JH, Rabelink TJ, van Bockel JH. Immunosuppression and the
abdominal aortic aneurysm: Doctor Jekyll or Mister Hyde? Circulation.
2011;124:e463–e465. doi: 10.1161/CIRCULATIONAHA.110.008573
178. Gallagher KA, Ravin RA, Schweitzer E, Stern T, Bartlett ST. Outcomes
and timing of aortic surgery in renal transplant patients. Ann Vasc Surg.
2011;25:448–453. doi: 10.1016/j.avsg.2010.12.007
179. Hoogduijn MJ, Crop MJ, Korevaar SS, Peeters AM, Eijken M, Maat LP,
Balk AH, Weimar W, Baan CC. Susceptibility of human mesenchymal stem
cells to tacrolimus, mycophenolic acid, and rapamycin. Transplantation.
2008;86:1283–1291. doi: 10.1097/TP.0b013e31818aa536
180. Leopardi M, Di Marco E, Musilli A, Ricevuto E, Bruera G, Ventura M.
Effects of chemotherapy in patients with concomitant aortic aneurysm
and malignant disease. Ann Vasc Surg. 2017;45:268.e13–268.e20. doi:
10.1016/j.avsg.2017.07.013
181. De Francesco EM, Bonuccelli G, Maggiolini M, Sotgia F, Lisanti MP.
Vitamin C and doxycycline: a synthetic lethal combination therapy tar-
geting metabolic flexibility in cancer stem cells (CSCs). Oncotarget.
2017;8:67269–67286. doi: 10.18632/oncotarget.18428
182. Lindeman JH, Ashcroft BA, Beenakker JW, van Es M, Koekkoek NB,
Prins FA, Tielemans JF, Abdul-Hussien H, Bank RA, Oosterkamp
TH. Distinct defects in collagen microarchitecture underlie vessel-
wall failure in advanced abdominal aneurysms and aneurysms in
Marfan syndrome. Proc Natl Acad Sci USA. 2010;107:862–865. doi:
10.1073/pnas.0910312107
183. Doderer SA, Gäbel G, Kokje VBC, Northoff BH, Holdt LM, Hamming
JF, Lindeman JHN. Adventitial adipogenic degeneration is an unidentified
contributor to aortic wall weakening in the abdominal aortic aneurysm. J
Vasc Surg. 2018;67:1891.e4–1900.e4. doi: 10.1016/j.jvs.2017.05.088
184. Kugo H, Zaima N, Tanaka H, Mouri Y, Yanagimoto K, Hayamizu K,
Hashimoto K, Sasaki T, Sano M, Yata T, Urano T, Setou M, Unno N,
Moriyama T. Adipocyte in vascular wall can induce the rupture of abdom-
inal aortic aneurysm. Sci Rep. 2016;6:31268. doi: 10.1038/srep31268
185. Birbrair A, Zhang T, Wang ZM, Messi ML, Enikolopov GN, Mintz
A, Delbono O. Role of pericytes in skeletal muscle regeneration
Downloaded from http://ahajournals.org by on February 20, 2019
Lindeman and Matsumura Pharmacologic Management of Aneurysms 645
and fat accumulation. Stem Cells Dev. 2013;22:2298–2314. doi:
10.1089/scd.2012.0647
186. Joe AW, Yi L, Natarajan A, Le Grand F, So L, Wang J, Rudnicki MA,
Rossi FM. Muscle injury activates resident fibro/adipogenic progeni-
tors that facilitate myogenesis. Nat Cell Biol. 2010;12:153–163. doi:
10.1038/ncb2015
187. Gäbel G, Northoff BH, Weinzierl I, Ludwig S, Hinterseher I, Wilfert W,
Teupser D, Doderer SA, Bergert H, Schönleben F, Lindeman JHN, Holdt
LM. Molecular fingerprint for terminal abdominal aortic aneurysm dis-
ease. J Am Heart Assoc. 2017;6:e006798.
188. Domergue S, Jorgensen C, Noël D. Advances in research in ani-
mal models of burn-related hypertrophic scarring. J Burn Care Res.
2015;36:e259–e266. doi: 10.1097/BCR.0000000000000167
189. Tashiro J, Rubio GA, Limper AH, Williams K, Elliot SJ, Ninou I, Aidinis
V, Tzouvelekis A, Glassberg MK. Exploring animal models that resem-
ble idiopathic pulmonary fibrosis. Front Med (Lausanne). 2017;4:118.
doi: 10.3389/fmed.2017.00118
190. Maegdefessel L, Azuma J, Toh R, Merk DR, Deng A, Chin JT, Raaz U,
Schoelmerich AM, Raiesdana A, Leeper NJ, McConnell MV, Dalman
RL, Spin JM, Tsao PS. Inhibition of microRNA-29b reduces murine ab-
dominal aortic aneurysm development. J Clin Invest. 2012;122:497–506.
doi: 10.1172/JCI61598
191. Rowbotham SE, Cavaye D, Jaeggi R, Jenkins JS, Moran CS, Moxon
JV, Pinchbeck JL, Quigley F, Reid CM, Golledge J. Fenofibrate in
the management of AbdoMinal aortic anEurysm (FAME): study
protocol for a randomised controlled trial. Trials. 2017;18:1. doi:
10.1186/s13063-016-1752-z
192. Rowbotham SE, Pinchbeck JL, Anderson G, Bourke B, Bourke M,
Gasser TC, Jaeggi R, Jenkins JS, Moran CS, Morton SK, Reid CM, Velu
R, Yip L, Moxon JV, Golledge J. Inositol in the MAnaGemENt of ab-
dominal aortic aneurysm (IMAGEN): study protocol for a randomised
controlled trial. Trials. 2017;18:547. doi: 10.1186/s13063-017-2304-x
193. Wang SK, Green LA, Gutwein AR, Drucker NA, Motaganahalli RL,
Fajardo A, Babbey CM, Murphy MP. Rationale and design of the
ARREST trial investigating mesenchymal stem cells in the treatment of
small abdominal aortic aneurysm. Ann Vasc Surg. 2018;47:230–237. doi:
10.1016/j.avsg.2017.08.044
194. Groth KA, Von Kodolitsch Y, Kutsche K, Gaustadnes M, Thorsen K,
Andersen NH, Gravholt CH. Evaluating the quality of Marfan geno-
type-phenotype correlations in existing FBN1 databases. Genet Med.
2017;19:772–777. doi: 10.1038/gim.2016.181
195. Sakai LY, Keene DR, Renard M, De Backer J. FBN1: the disease-
causing gene for Marfan syndrome and other genetic disorders. Gene.
2016;591:279–291. doi: 10.1016/j.gene.2016.07.033
196. Loeys BL, Dietz HC, Braverman AC, Callewaert BL, De Backer J,
Devereux RB, Hilhorst-Hofstee Y, Jondeau G, Faivre L, Milewicz DM,
Pyeritz RE, Sponseller PD, Wordsworth P, De Paepe AM. The revised
Ghent nosology for the Marfan syndrome. J Med Genet. 2010;47:476–
485. doi: 10.1136/jmg.2009.072785
197. von Kodolitsch Y, Spielmann RP, Nienaber CA. Acute and chronic aortic
diseases in Marfan syndrome and arterial hypertension–a comparison of
anatomy, clinical aspects and prognosis. Z Kardiol. 1995;84:542–552.
198. Engelfriet PM, Boersma E, Tijssen JG, Bouma BJ, Mulder BJ. Beyond
the root: dilatation of the distal aorta in Marfan’s syndrome. Heart.
2006;92:1238–1243. doi: 10.1136/hrt.2005.081638
199. Zeyer KA, Reinhardt DP. Fibrillin-containing microfibrils are key signal
relay stations for cell function. J Cell Commun Signal. 2015;9:309–325.
doi: 10.1007/s12079-015-0307-5
200. Gray JR, Bridges AB, West RR, McLeish L, Stuart AG, Dean JC,
Porteous ME, Boxer M, Davies SJ. Life expectancy in British Marfan
syndrome populations. Clin Genet. 1998;54:124–128.
201. Krause KJ. Marfan syndrome: literature review of mortality studies. J
Insur Med. 2000;32:79–88.
202. Franken R, Groenink M, de Waard V, Feenstra HM, Scholte AJ, van den
Berg MP, Pals G, Zwinderman AH, Timmermans J, Mulder BJ. Genotype
impacts survival in Marfan syndrome. Eur Heart J. 2016;37:3285–3290.
doi: 10.1093/eurheartj/ehv739
203. Franken R, Teixido-Tura G, Brion M, Forteza A, Rodriguez-Palomares
J, Gutierrez L, Garcia Dorado D, Pals G, Mulder BJ, Evangelista A.
Relationship between fibrillin-1 genotype and severity of cardiovascular
involvement in Marfan syndrome. Heart. 2017;103:1795–1799. doi:
10.1136/heartjnl-2016-310631
204. Erbel R, Aboyans V, Boileau C, et al; ESC Committee for Practice
Guidelines. 2014 ESC Guidelines on the diagnosis and treatment of
aortic diseases: document covering acute and chronic aortic diseases
of the thoracic and abdominal aorta of the adult. The Task Force for
the Diagnosis and Treatment of Aortic Diseases of the European
Society of Cardiology (ESC). Eur Heart J. 2014;35:2873–2926. doi:
10.1093/eurheartj/ehu281
205. Shores J, Berger KR, Murphy EA, Pyeritz RE. Progression of aortic
dilatation and the benefit of long-term beta-adrenergic blockade
in Marfan’s syndrome. N Engl J Med. 1994;330:1335–1341. doi:
10.1056/NEJM199405123301902
206. Chun AS, Elefteriades JA, Mukherjee SK. Do β-blockers really work
for prevention of aortic aneurysms?: time for reassessment. Aorta
(Stamford). 2013;1:45–51. doi: 10.12945/j.aorta.2013.13.002
207. Silverman DI, Burton KJ, Gray J, Bosner MS, Kouchoukos NT, Roman
MJ, Boxer M, Devereux RB, Tsipouras P. Life expectancy in the Marfan
syndrome. Am J Cardiol. 1995;75:157–160.
208. Salim MA, Alpert BS, Ward JC, Pyeritz RE. Effect of beta-adrenergic
blockade on aortic root rate of dilation in the Marfan syndrome. Am J
Cardiol. 1994;74:629–633.
209. Ladouceur M, Fermanian C, Lupoglazoff JM, Edouard T, Dulac Y,
Acar P, Magnier S, Jondeau G. Effect of beta-blockade on ascending
aortic dilatation in children with the Marfan syndrome. Am J Cardiol.
2007;99:406–409. doi: 10.1016/j.amjcard.2006.08.048
210. Selamet Tierney ES, Feingold B, Printz BF, Park SC, Graham D,
Kleinman CS, Mahnke CB, Timchak DM, Neches WH, Gersony WM.
Beta-blocker therapy does not alter the rate of aortic root dilation in pe-
diatric patients with Marfan syndrome. J Pediatr. 2007;150:77–82. doi:
10.1016/j.jpeds.2006.09.003
211. Yetman AT, Bornemeier RA, McCrindle BW. Usefulness of enalapril
versus propranolol or atenolol for prevention of aortic dilation in patients
with the Marfan syndrome. Am J Cardiol. 2005;95:1125–1127. doi:
10.1016/j.amjcard.2005.01.032
212. Rossi-Foulkes R, Roman MJ, Rosen SE, Kramer-Fox R, Ehlers KH,
O’Loughlin JE, Davis JG, Devereux RB. Phenotypic features and impact
of beta blocker or calcium antagonist therapy on aortic lumen size in the
Marfan syndrome. Am J Cardiol. 1999;83:1364–1368.
213. Fitzmaurice GM, Ravichandran C. A primer in longitudinal data analysis.
Circulation. 2008;118:2005–2010. doi: 10.1161/CIRCULATIONAHA.
107.714618
214. Engelfriet P, Mulder B. Is there benefit of beta-blocking agents in
the treatment of patients with the Marfan syndrome? Int J Cardiol.
2007;114:300–302. doi: 10.1016/j.ijcard.2006.01.025
215. Chun AS, Elefteriades JA, Mukherjee SK. Do β-blockers really work
for prevention of aortic aneurysms?: time for reassessment. Aorta
(Stamford). 2013;1:45–51. doi: 10.12945/j.aorta.2013.13.002
216. Koo HK, Lawrence KA, Musini VM. Beta-blockers for preventing
aortic dissection in Marfan syndrome. Cochrane Database Syst Rev.
2017;11:CD011103. doi: 10.1002/14651858.CD011103.pub2
217. Rios AS, Silber EN, Bavishi N, Varga P, Burton BK, Clark WA, Denes P.
Effect of long-term beta-blockade on aortic root compliance in patients
with Marfan syndrome. Am Heart J. 1999;137:1057–1061.
218. Haouzi A, Berglund H, Pelikan PC, Maurer G, Siegel RJ. Heterogeneous
aortic response to acute beta-adrenergic blockade in Marfan syndrome.
Am Heart J. 1997;133:60–63.
219. Habashi JP, Judge DP, Holm TM, et al. Losartan, an AT1 antagonist, pre-
vents aortic aneurysm in a mouse model of Marfan syndrome. Science.
2006;312:117–121. doi: 10.1126/science.1124287
220. Chiu HH, Wu MH, Wang JK, Lu CW, Chiu SN, Chen CA, Lin MT,
Hu FC. Losartan added to β-blockade therapy for aortic root dilation
in Marfan syndrome: a randomized, open-label pilot study. Mayo Clin
Proc. 2013;88:271–276. doi: 10.1016/j.mayocp.2012.11.005
221. Pees C, Laccone F, Hagl M, Debrauwer V, Moser E, Michel-
Behnke I. Usefulness of losartan on the size of the ascending aorta
in an unselected cohort of children, adolescents, and young adults
with Marfan syndrome. Am J Cardiol. 2013;112:1477–1483. doi:
10.1016/j.amjcard.2013.06.019
222. Brooke BS, Habashi JP, Judge DP, Patel N, Loeys B, Dietz HC III.
Angiotensin II blockade and aortic-root dilation in Marfan’s syndrome.
N Engl J Med. 2008;358:2787–2795. doi: 10.1056/NEJMoa0706585
223. Sandor GG, Alghamdi MH, Raffin LA, Potts MT, Williams LD, Potts
JE, Kiess M, van Breemen C. A randomized, double blind pilot study to
assess the effects of losartan vs. atenolol on the biophysical properties
of the aorta in patients with Marfan and Loeys-Dietz syndromes. Int J
Cardiol. 2015;179:470–475. doi: 10.1016/j.ijcard.2014.11.082
224. Muiño-Mosquera L, De Nobele S, Devos D, Campens L, De Paepe A,
De Backer J. Efficacy of losartan as add-on therapy to prevent aortic
growth and ventricular dysfunction in patients with Marfan syndrome: a
Downloaded from http://ahajournals.org by on February 20, 2019
646 Circulation Research February 15, 2019
randomized, double-blind clinical trial. Acta Cardiol. 2017;72:616–624.
doi: 10.1080/00015385.2017.1314134
225. Forteza A, Evangelista A, Sánchez V, Teixidó-Turà G, Sanz P, Gutiérrez
L, Gracia T, Centeno J, Rodríguez-Palomares J, Rufilanchas JJ,
Cortina J, Ferreira-González I, García-Dorado D. Efficacy of losar-
tan vs. atenolol for the prevention of aortic dilation in Marfan syn-
drome: a randomized clinical trial. Eur Heart J. 2016;37:978–985. doi:
10.1093/eurheartj/ehv575
226. Milleron O, Arnoult F, Ropers J, et al. Marfan Sartan: a randomized,
double-blind, placebo-controlled trial. Eur Heart J. 2015;36:2160–2166.
doi: 10.1093/eurheartj/ehv151
227. Lacro RV, Dietz HC, Sleeper LA, et al; Pediatric Heart Network
Investigators. Atenolol versus losartan in children and young adults
with Marfan’s syndrome. N Engl J Med. 2014;371:2061–2071. doi:
10.1056/NEJMoa1404731
228. Groenink M, den Hartog AW, Franken R, Radonic T, de Waard V,
Timmermans J, Scholte AJ, van den Berg MP, Spijkerboer AM,
Marquering HA, Zwinderman AH, Mulder BJ. Losartan reduces aortic
dilatation rate in adults with Marfan syndrome: a randomized controlled
trial. Eur Heart J. 2013;34:3491–3500. doi: 10.1093/eurheartj/eht334
229. Franken R, den Hartog AW, Radonic T, Micha D, Maugeri A, van Dijk
FS, Meijers-Heijboer HE, Timmermans J, Scholte AJ, van den Berg
MP, Groenink M, Mulder BJ, Zwinderman AH, de Waard V, Pals G.
Beneficial outcome of losartan therapy depends on type of FBN1 muta-
tion in Marfan syndrome. Circ Cardiovasc Genet. 2015;8:383–388. doi:
10.1161/CIRCGENETICS.114.000950
230. Forfar JC. A randomised, double-blind, placebo-controlled pilot trial of irbe-
sartan, doxycycline and a combination on markers of vascular dysfunction
in the Marfan syndrome, using cardiovascular magnetic resonance imaging.
https://www.clinicaltrialsregister.eu/ctr-search/search?query=eudract_
number:2010-023612-14. Accessed January 2, 2019.
231. Gao L, Chen L, Fan L, Gao D, Liang Z, Wang R, Lu W. The effect of
losartan on progressive aortic dilatation in patients with Marfan’s syn-
drome: a meta-analysis of prospective randomized clinical trials. Int J
Cardiol. 2016;217:190–194. doi: 10.1016/j.ijcard.2016.04.186
232. Meijboom LJ, Timmermans J, Zwinderman AH, Engelfriet PM, Mulder
BJ. Aortic root growth in men and women with the Marfan’s syndrome.
Am J Cardiol. 2005;96:1441–1444. doi: 10.1016/j.amjcard.2005.06.094
233. Cavanaugh NB, Qian L, Westergaard NM, Kutschke WJ, Born EJ,
Turek JW. A novel murine model of Marfan syndrome accelerates aor-
topathy and cardiomyopathy. Ann Thorac Surg. 2017;104:657–665. doi:
10.1016/j.athoracsur.2016.10.077
234. Chen M, Yao B, Yang Q, Deng J, Song Y, Sui T, Zhou L, Yao H, Xu
Y, Ouyang H, Pang D, Li Z, Lai L. Truncated C-terminus of fibrillin-1
induces Marfanoid-progeroid-lipodystrophy (MPL) syndrome in rabbit.
Dis Model Mech. 2018;11:dmm031542.
235. Cook JR, Clayton NP, Carta L, Galatioto J, Chiu E, Smaldone S,
Nelson CA, Cheng SH, Wentworth BM, Ramirez F. Dimorphic
effects of transforming growth factor-β signaling during aortic aneu-
rysm progression in mice suggest a combinatorial therapy for Marfan
syndrome. Arterioscler Thromb Vasc Biol. 2015;35:911–917. doi:
10.1161/ATVBAHA.114.305150
236. Curtis AE, Smith TA, Ziganshin BA, Elefteriades JA. The mystery of
the Z-score. Aorta (Stamford). 2016;4:124–130. doi: 10.12945/j.aorta.
2016.16.014
237. Pitcher A, Emberson J, Lacro RV, et al. Design and rationale of a prospec-
tive, collaborative meta-analysis of all randomized controlled trials of an-
giotensin receptor antagonists in Marfan syndrome, based on individual
patient data: a report from the Marfan Treatment Trialists’ Collaboration.
Am Heart J. 2015;169:605–612. doi: 10.1016/j.ahj.2015.01.011
238. Milewicz DM, Prakash SK, Ramirez F. Therapeutics targeting drivers
of thoracic aortic aneurysms and acute aortic dissections: insights from
predisposing genes and mouse models. Annu Rev Med. 2017;68:51–67.
doi: 10.1146/annurev-med-100415-022956
239. Franken R, Radonic T, den Hartog AW, Groenink M, Pals G,
van Eijk M, Lutter R, Mulder BJ, Zwinderman AH, de Waard V;
COMPARE Study Group. The revised role of TGF-β in aortic aneu-
rysms in Marfan syndrome. Neth Heart J. 2015;23:116–121. doi:
10.1007/s12471-014-0622-0
240. Wei H, Hu JH, Angelov SN, Fox K, Yan J, Enstrom R, Smith A, Dichek
DA. Aortopathy in a mouse model of Marfan syndrome is not mediated
by altered transforming growth factor β signaling. J Am Heart Assoc.
2017;6:e004968.
241. Dale M, Fitzgerald MP, Liu Z, et al. Premature aortic smooth muscle cell
differentiation contributes to matrix dysregulation in Marfan Syndrome.
PLoS One. 2017;12:e0186603. doi: 10.1371/journal.pone.0186603
242. Granata A, Serrano F, Bernard WG, McNamara M, Low L, Sastry P,
Sinha S. An iPSC-derived vascular model of Marfan syndrome identifies
key mediators of smooth muscle cell death. Nat Genet. 2017;49:97–109.
doi: 10.1038/ng.3723
243. Mallat Z, Daugherty A. AT1 receptor antagonism to reduce aortic ex-
pansion in Marfan syndrome: lost in translation or in need of different
interpretation? Arterioscler Thromb Vasc Biol. 2015;35:e10–e12. doi:
10.1161/ATVBAHA.114.305173
244. Van Wijk BL, Klungel OH, Heerdink ER, de Boer A. Rate and determi-
nants of 10-year persistence with antihypertensive drugs. J Hypertens.
2005;23:2101–2107.
245. Teixido-Tura G, Forteza A, Rodríguez-Palomares J, González Mirelis
J, Gutiérrez L, Sánchez V, Ibáñez B, García-Dorado D, Evangelista A.
Losartan versus atenolol for prevention of aortic dilation in patients
with Marfansyndrome. J Am Coll Cardiol. 2018;72:1613–1618. doi:
10.1016/j.jacc.2018.07.052
Downloaded from http://ahajournals.org by on February 20, 2019
... AAAs represent a heterogenous group of disorders dependent on the location of the aneurysm, type of dilatation, size of the aneurysm, and complications (16,17,31,(37)(38)(39). In general, the prevalence of AAA ranges from 4-9% with significant increases in those >65 years old, male gender, and smokers (17,37,39). ...
... AAAs represent a heterogenous group of disorders dependent on the location of the aneurysm, type of dilatation, size of the aneurysm, and complications (16,17,31,(37)(38)(39). In general, the prevalence of AAA ranges from 4-9% with significant increases in those >65 years old, male gender, and smokers (17,37,39). The most feared complication is aneurysmal rupture with a resulting mortality ranging from 50-90% (37,39). ...
... In general, the prevalence of AAA ranges from 4-9% with significant increases in those >65 years old, male gender, and smokers (17,37,39). The most feared complication is aneurysmal rupture with a resulting mortality ranging from 50-90% (37,39). The pathophysiology of the development and expansion of AAAs is still debated but thought to be secondary to localized inflammation rather than poorly controlled blood pressure (39). ...
... Currently, no medical treatment is available and over a dozen clinical trials on early AAA growth abrogation have failed to translate in vitro results into clinical success. [5,6] Additionally, secondary complications such as early or late EVAR failure or suture aneurysm after OAR due to dilated sealing zones or disease progression are not infrequent. [7] Here, two shortcomings must be considered: a still incomplete understanding of the AAA pathogenesis and the possible heterogeneity of disease among individuals. ...
... Thus medical growth abrogation as suggested and partially tested by others and us, might only be applicable to specific patient populations. [5,27,28] Vice versa, smoking cessation, as suggested by international guidelines, associated with reduced aneurysm growth was found to be linked to more chronic inflammation and more fibrosis suggestive of higher inflammatory activity. [1,3,4,29] Targeting specific subtypes of inflammation in AAA is shaping up to be an interesting field of future research, especially in regards to targeted therapies. ...
Preprint
Objective: Abdominal aortic aneurysm (AAA) treatment is upon a diameter threshold by open (OAR) or endovascular aortic repair. Attempts for medical growth abrogation have failed. This study aims to elucidate the heterogeneity of AAA based on histomorphology in correlation to individual patient data and aneurysm metrics. Patients and Methods: Aneurysm samples from the left anterior wall from four university center biobanks underwent histologic analysis including angiogenesis, calcification, fibrosis, type and grade of inflammation in adventitia and media. Clinical information included age, comorbidities, etc. as well as type of aneurysm (intact, symptomatic, ruptured, inflammatory). Aneurysm morphology included diameter and semi-automated geometric analysis using Endosize (Therenva) segmentation. Additionally, aneurysm growth was assessed. Results: 364 patient samples (85.4% male, median age 69 years) demonstrated acute (mix/granulocytes) or chronic (monocytes/plasma cells) type inflammation and score, coherent in adventitia and media (p < 0.001), not associated with the type of aneurysm (52x ruptured; 37x symptomatic; p = 0.51) or diameter (57 [52-69] mm; p = 0.87). The degree of fibrosis and the presence of angiogenesis were significantly higher (both p < 0.001) with increasing inflammation score, which significantly decreased with patient age (est = - 0.015/year, p = 0.017). No significant differences in inflammation, fibrosis or angiogenesis were seen for ruptured (vs. intact), acute (vs. elective), male (vs. female) of diabetic (vs. non) patients, while current smoking was associated with more chronic inflammation (p = 0.007) and a higher degree of fibrosis (p = 0.03). Aneurysm geometric morphology (n=252) or differing annual growth rates (n=142) were not significantly associated with histologic characteristics. Conclusion: Type and degree of inflammation are the most distinguishable histologic characteristics in the AAA wall between individual patients. Despite the association to age and smoking status, no significant correlation to any patient or aneurysm specific feature, especially not diameter or rupture exists.
... While surgery is the definitive strategy for preventing AAA rupture [1], it provides no therapeutic advantage to patients with small AAAs or surgical contraindications. Despite numerous experimental and clinical studies aiming to develop effective medical therapies for AAA, no pharmaceutical treatment is currently available to arrest or limit AAA growth or to prevent aortic rupture [3]. Therefore, the management of AAA remains clinically challenging. ...
Article
Full-text available
Abdominal aortic aneurysm (AAA) is a chronic vascular degenerative disease characterized by progressive segmental dilation of the abdominal aorta. The rupture of an AAA represents a leading cause of death in cardiovascular diseases. Despite numerous experimental and clinical studies examining potential drug targets and therapies, currently there are no pharmaceutical treatment to prevent AAA growth and rupture. Iron is an essential element in almost all living organisms and has important biological functions. Epidemiological studies have indicated that both iron deficiency and overload are associated with adverse clinical outcomes, particularly an increased risk of cardiovascular events. Recent evidence indicates that iron overload is involved in the pathogenesis of abdominal aortic aneurysms. In this review, we provide an overview of the role of iron overload in AAA progression and explore its potential pathological mechanisms. Although the exact molecular mechanisms of iron overload in the development of AAA remain to be elucidated, the inhibition of iron deposition may offer a promising strategy for preventing these aneurysms.
... Clinically, most AAAs are asymptomatic and are often detected as incidental findings during the investigation of an unrelated problem or during radiological screening (52)(53)(54). Multiple trials have shown no benefit of repairing AAA that is less than 55 mm in diameter, and consequently, current guidelines advise watchful waiting for aneurysms less than 55 mm and repair once the AAA grows to greater than 55 mm (55). Therefore, further study is required to evaluate the potential of IGF1C-loaded hydrogels to slow or halt the progression of aneurysms between 30 and 55 mm in size. ...
Article
Full-text available
An abdominal aortic aneurysm (AAA) is a life-threatening cardiovascular disease. We identified plasma insulin-like growth factor 1 (IGF1) as an independent risk factor in patients with AAA by correlating plasma IGF1 with risk. Smooth muscle cell– or fibroblast-specific knockout of Igf1r , the gene encoding the IGF1 receptor (IGF1R), attenuated AAA formation in two mouse models of AAA induced by angiotensin II infusion or CaCl 2 treatment. IGF1R was activated in aortic aneurysm samples from human patients and mice with AAA. Systemic administration of IGF1C, a peptide fragment of IGF1, 2 weeks after disease development inhibited AAA progression in mice. Decreased AAA formation was linked to competitive inhibition of IGF1 binding to its receptor by IGF1C and modulation of downstream alpha serine/threonine protein kinase (AKT)/mammalian target of rapamycin signaling. Localized application of an IGF1C-loaded hydrogel was developed to reduce the side effects observed after systemic administration of IGF1C or IGF1R antagonists in the CaCl 2 -induced AAA mouse model. The inhibitory effect of the IGF1C-loaded hydrogel administered at disease onset on AAA formation was further evaluated in a guinea pig-to-rat xenograft model and in a sheep-to-minipig xenograft model of AAA formation. The therapeutic efficacy of IGF1C for treating AAA was tested through extravascular delivery in the sheep-to-minipig model with AAA established for 2 weeks. Percutaneous injection of the IGF1C-loaded hydrogel around the AAA resulted in improved vessel flow dynamics in the minipig aorta. These findings suggest that extravascular administration of IGF1R antagonists may have translational potential for treating AAA.
Article
The treatment of abdominal aortic aneurysms (AAA) depends on the diameter and is primarily performed as surgical or endovascular repair. In recent years interventional studies on conservative stabilization of aneurysms failed to show a positive effect. Based on a systematic presentation previous studies on the topic are discussed and potential limitations are shown. Based on a scoping review the authors present their own research approaches on the molecular pathogenesis of AAA. Due to the heterogeneous pathobiology a patient-specific consideration is necessary. In the past the treatment with sartans, doxycycline, macrolides, propranolol, statins and angiotensin-converting enzyme (ACE) inhibitors did not lead to a relevant reduction in aneurysm growth; however, there is promising evidence from the use of metformin in diabetic patients with AAA. Furthermore, drugs with pleiotropic effects have shown promising results in small and large animal models as well as approaches for targeted inhibition of platelet function. Stabilization of the aneurysm by local treatment with non-coding RNA equivalents or an endovascular device in the aneurysm neck with a so-called circumferential aortic stiffening device (CASD) also showed a preclinical reduction of the increase in diameter. Drug trials on controlling AAA progression must take patient-specific pathophysiological aspects, a sufficient number of patients and duration of follow-up into account. Several approaches for drug-based AAA treatment are currently being pursued with agents already in clinical use for other indications. Clinical trials on metformin in non-diabetic patients with AAA are the most advanced.
Article
BACKGROUND Abdominal aortic aneurysm (AAA) is a chronic vascular inflammatory disease without effective medications. PCSK9 (proprotein convertase subtilisin/kexin 9), a serine protease from the proprotein convertase family, has recently been associated with AAA in human genome-wide association studies. However, its role in AAA is unknown. METHODS Transcriptional and histological expression of PCSK9 was examined in AAA tissues and healthy controls. The impact of PCSK9 deletion and inhibition on AAA formation was assessed in mice with hyperlipidemia and Ang II (angiotensin II) overproduction. AAA lesion morphology was assessed by tissue staining. MMP (matrix metalloproteinase) activity was evaluated by gelatin zymography, and leukocyte-vessel wall interaction was monitored by intravital microscopy. RNA sequencing was used to characterize the downstream signaling of PCSK9. RESULTS PCSK9 expression was upregulated and colocalized with macrophages in human and mouse AAAs. Pcsk9 deletion attenuated AAA formation, improved survival, and decreased systemic inflammation, without altering circulating cholesterol levels. Pcsk9 deficiency reduced aortic infiltration of macrophages and elastin degradation, without affecting vascular smooth muscle cell apoptosis and proliferation. Mechanistically, PCSK9 was essential in leukocyte-endothelium adhesion and expression of proinflammatory cytokines and MMP9 by macrophages. RNA sequencing of stimulated macrophages revealed that Pcsk9 deficiency upregulated histone deacetylase SIRT1 (sirtuin-1) and suppressed NF-κB (nuclear factor-κB) inflammatory signaling. SIRT1 inhibition attenuated the proinflammatory actions of PCSK9. Furthermore, administration of PCSK9 small interfering RNA or antibody constrained AAA formation/progression and inhibited vascular inflammation. CONCLUSIONS PCSK9 critically mediates macrophage inflammation and elastin degradation, promoting AAA formation. PCSK9 inhibitors bear a promise to curtail AAA, beyond being used as cholesterol-lowering drugs.
Article
Aortic aneurysm (AA) refers to the persistent dilatation of the aorta, exceeding three centimeters. Investigating the pathophysiology of this condition is important for its prevention and management, given its responsibility for more than 25000 deaths in the United States. AAs are classified based on their location or morphology. various pathophysiologic pathways including inflammation, the immune system and atherosclerosis have been implicated in its development. Inflammatory markers such as transforming growth factor β, interleukin-1β, tumor necrosis factor-α, matrix metalloproteinase-2 and many more may contribute to this phenomenon. Several genetic disorders such as Marfan syndrome, Ehler-Danlos syndrome and Loeys-Dietz syndrome have also been associated with this disease. Recent years has seen the investigation of novel management of AA, exploring the implication of different immune suppressors, the role of radiation in shrinkage and prevention, as well as minimally invasive and newly hypothesized surgical methods. In this narrative review, we aim to present the new contributing factors involved in pathophysiology of AA. We also highlighted the novel management methods that have demonstrated promising benefits in clinical outcomes of the AA.
Article
Full-text available
Abdominal Aortic Aneurysm (AAA) is a disease characterized by localized dilation of the abdominal aorta, involving multiple factors in its occurrence and development, ultimately leading to vessel rupture and severe bleeding. AAA has a high mortality rate, and there is a lack of targeted therapeutic drugs. Epigenetic regulation plays a crucial role in AAA, and the treatment of AAA in the epigenetic field may involve a series of related genes and pathways. Abnormal expression of these genes may be a key factor in the occurrence of the disease and could potentially serve as promising therapeutic targets. Understanding the epigenetic regulation of AAA is of significant importance in revealing the mechanisms underlying the disease and identifying new therapeutic targets. This knowledge can contribute to offering AAA patients better clinical treatment options beyond surgery. This review systematically explores various aspects of epigenetic regulation in AAA, including DNA methylation, histone modification, non-coding RNA, and RNA modification. The analysis of the roles of these regulatory mechanisms, along with the identification of relevant genes and pathways associated with AAA, is discussed comprehensively. Additionally, a comprehensive discussion is provided on existing treatment strategies and prospects for epigenetics-based treatments, offering insights for future clinical interventions.
Article
Full-text available
The article analyzed the results of surgical treatment of 140 patients with surgery of abdominal aortic aneurism. The comparison group consisted of 80 patients with aortic aneurism more than 4,5 cm, who didn’t undergo surgery. The conventional method of Khardi-Pokrovskiy resection was complemented by a number of surgical methods in these cases. The results of surgery were improved due to application of these methods. All the patients (100%), who didn’t undergo surgery, passed away during 5 years, though 70% of them died because of aneurysm rupture. The early postoperative lethality was 5% in the main group, but 5-year survival was 81%.
Article
Full-text available
Objectives: In the course of extensive clinical aortic surgery, we noticed that the aorta was quite thick and fibrotic in diabetic patients. We thought the diabetic aortic aorta might be inimitable to aortic dissection. On this basis, we set out to review information in the literature regarding aortic growth and dissection in diabetic patients. Methods: We used a 2-step search approach to the available literature on diabetes and aneurysm. Firstly, databases including PubMed, Cochrane, Embase and TRIP were searched. Secondly, relevant studies were identified through secondary sources including references of initially selected articles. We address the relationship between diabetes and the incidence, prevalence, growth, mortality and rupture of an aneurysm. Results: Diabetes is thought to exert a protective role in both thoracic aortic aneurysm (TAA) and abdominal aortic aneurysm (AAA). Diabetics were shown to have a slower aneurysm growth rate, lower rupture rate, delayed (> 65 years) age of rupture, decreased rate of mortality from an aneurysm and a decreased length of hospital stay. There was also noted a decreased rate of incidence and prevalence of TAA and AAA in diabetics, smaller aneurysm diameter, reduction in matrix metalloproteinases and an increased aortic wall stress in diabetics. Antidiabetic agents like metformin, thiazolidinediones and dipeptidyl peptidase-4 inhibitors may protect against an aneurysm. Conclusion: Our literature review provides strong (but often circumstantial) evidence that diabetic patients exhibit slower growth of aortic aneurysms and a lower rate of aortic dissection. Furthermore, clinical and experimental studies indicate that common antidiabetic medications on their own inhibit growth of aortic aneurysms. These findings indicate a paradoxically beneficial effect of the otherwise highly detrimental diabetic state.
Article
Full-text available
Background There is no drug therapy for abdominal aortic aneurysm (AAA). FAME‐2 (Fenofibrate in the Management of Abdominal Aortic Aneurysm 2) was a placebo‐controlled randomized trial designed to assess whether administration of 145 mg of fenofibrate/d for 24 weeks favorably modified circulating markers of AAA. Methods and Results Patients with AAAs measuring 35 to 49 mm and no contraindication were randomized to fenofibrate or identical placebo. The primary outcome measures were the differences in serum osteopontin and kallistatin concentrations between groups. Secondary analyses compared changes in the circulating concentration of AAA‐associated proteins, and AAA growth, between groups using multivariable linear mixed‐effects modeling. A total of 140 patients were randomized to receive fenofibrate (n=70) or placebo (n=70). By the end of the study 3 (2.1%) patients were lost to follow‐up and 18 (12.9%) patients had ceased trial medication. A total of 85% of randomized patients took ≥80% of allocated tablets and were deemed to have complied with the medication regimen. Patients’ allocated fenofibrate had expected reductions in serum triglycerides and estimated glomerular filtration rate, and increases in serum homocysteine. No differences in serum osteopontin, kallistatin, or AAA growth were observed between groups. Conclusions Administering 145 mg/d of fenofibrate for 24 weeks did not significantly reduce serum concentrations of osteopontin and kallistatin concentrations, or rates of AAA growth in this trial. The findings do not support the likely benefit of fenofibrate as a treatment for patients with small AAAs. Clinical Trial Registration URL: http://www.anzctr.org.au. Unique identifier: ACTRN12613001039774.
Article
Endovascular aneurysm repair (EVAR) has become the preferred strategy for elective repair of abdominal aortic aneurysm (AAA) for many patients. However, the superiority of the endovascular procedure has recently been challenged by reports of impaired long-term survival in patients who underwent EVAR. A systematic review of long-term survival following AAA repair was therefore undertaken. METHODS: A systematic review was performed according to PRISMA guidelines. Articles reporting short- and/or long-term mortality of EVAR and open surgical repair (OSR) of AAA were identified. Pooled overall survival estimates (hazard ratios (HRs) with corresponding 95 per cent c.i. for EVAR versus OSR) were calculated using a random-effects model. Possible confounding owing to age differences between patients receiving EVAR or OSR was addressed by estimating relative survival. RESULTS: Some 53 studies were identified. The 30-day mortality rate was lower for EVAR compared with OSR: 1·16 (95 per cent c.i. 0·92 to 1·39) versus 3·27 (2·71 to 3·83) per cent. Long-term survival rates were similar for EVAR versus OSR (HRs 1·01, 1·00 and 0·98 for 3, 5 and 10 years respectively; P = 0·721, P = 0·912 and P = 0·777). Correction of age inequality by means of relative survival analysis showed equal long-term survival: 0·94, 0·91 and 0·76 at 3, 5 and 10 years for EVAR, and 0·96, 0·91 and 0·76 respectively for OSR. CONCLUSION: Long-term overall survival rates were similar for EVAR and OSR. Available data do not allow extension beyond the 10-year survival window or analysis of specific subgroups.
Article
Background Endovascular aneurysm repair (EVAR) has become the preferred strategy for elective repair of abdominal aortic aneurysm (AAA) for many patients. However, the superiority of the endovascular procedure has recently been challenged by reports of impaired long‐term survival in patients who underwent EVAR. A systematic review of long‐term survival following AAA repair was therefore undertaken. Methods A systematic review was performed according to PRISMA guidelines. Articles reporting short‐ and/or long‐term mortality of EVAR and open surgical repair (OSR) of AAA were identified. Pooled overall survival estimates (hazard ratios (HRs) with corresponding 95 per cent c.i. for EVAR versus OSR) were calculated using a random‐effects model. Possible confounding owing to age differences between patients receiving EVAR or OSR was addressed by estimating relative survival. Results Some 53 studies were identified. The 30‐day mortality rate was lower for EVAR compared with OSR: 1·16 (95 per cent c.i. 0·92 to 1·39) versus 3·27 (2·71 to 3·83) per cent. Long‐term survival rates were similar for EVAR versus OSR (HRs 1·01, 1·00 and 0·98 for 3, 5 and 10 years respectively; P = 0·721, P = 0·912 and P = 0·777). Correction of age inequality by means of relative survival analysis showed equal long‐term survival: 0·94, 0·91 and 0·76 at 3, 5 and 10 years for EVAR, and 0·96, 0·91 and 0·76 respectively for OSR. Conclusion Long‐term overall survival rates were similar for EVAR and OSR. Available data do not allow extension beyond the 10‐year survival window or analysis of specific subgroups.
Article
Background: Beta-blockers are the standard treatment in Marfan syndrome (MFS). Recent clinical trials with limited follow-up yielded conflicting results on losartan's effectiveness in MFS. Objectives: The present study aimed to evaluate the benefit of losartan compared with atenolol for the prevention of aortic dilation and complications in Marfan patients over a longer observation period (>5 years). Methods: A total of 128 patients included in the previous LOAT (LOsartan vs ATenolol) clinical trial (64 in the atenolol and 64 in the losartan group) were followed up for an open-label extension of the study, with the initial treatment maintained. Results: Mean clinical follow-up was 6.7 ± 1.5 years. A total of 9 events (14.1%) occurred in the losartan group and 12 (18.8%) in the atenolol group. Survival analysis showed no differences in the combined endpoint of need for aortic surgery, aortic dissection, or death (p = 0.462). Aortic root diameter increased with no differences between groups: 0.4 mm/year (95% confidence interval: 0.2 to 0.5) in the losartan and 0.4 mm/year (95% confidence interval: 0.3 to 0.6) in the atenolol group. In the subgroup analyses, no significant differences were observed considering age, baseline aortic root diameter, or type of dominant negative versus haploinsufficient FBN1 mutation. Conclusions: Long-term outcome of Marfan syndrome patients randomly assigned to losartan or atenolol showed no differences in aortic dilation rate or presence of clinical events between treatment groups. Therefore, losartan might be a useful, low-risk alternative to beta-blockers in the long-term management of these patients.
Article
Background: Identification of a safe and effective medical therapy for abdominal aortic aneurysm (AAA) disease remains a significant unmet medical need. Recent small cohort studies indicate that metformin, the world's most commonly prescribed oral hypoglycemic agent, may limit AAA enlargement. We sought to validate these preliminary observations in a larger cohort. Methods: All patients with asymptomatic AAA disease managed in the Veterans Affairs Health Care System between 2003 and 2013 were identified by International Classification of Diseases, Ninth Revision codes. Those with a concomitant diagnosis of diabetes mellitus who also received two or more abdominal imaging studies (computed tomography, magnetic resonance imaging, or ultrasound) documenting the presence and size of an AAA, separated by at least 1 year, were included for review. Maximal AAA diameters were determined from radiologic reports. Further data acquisition was censored after surgical AAA repair, when performed. Comorbidities, active smoking status, and outpatient medication records (within 6 months of AAA diagnosis) were also queried. Yearly AAA enlargement rates, as a function of metformin treatment status, were compared using two statistical models expressed in millimeters per year: a multivariate linear regression (model 1) and a multivariate mixed-effects model with random intercept and random slope (model 2). Results: A total of 13,834 patients with 58,833 radiographic records were included in the analysis, with radiology imaging follow-up of 4.2 ± 2.6 years (mean ± standard deviation). The average age of the patients at AAA diagnosis was 69.8 ± 7.8 years, and 39.7% had a metformin prescription within ±6 months of AAA. The mean growth rate for AAAs in the entire cohort was 1.4 ± 2.0 mm/y by model 1 analysis and 1.3 ± 1.6 mm/y by model 2 analysis. The unadjusted mean rate of AAA growth was 1.2 ± 1.9 mm/y for patients prescribed metformin compared with 1.5 ± 2.2 mm/y for those without (P < .001), a 20% decrease. This effect remained significant when adjusted for variables relevant on AAA progression: metformin prescription was associated with a reduction in yearly AAA growth rate of -0.23 mm (95% confidence interval, -0.35 to -0.16; P < .001) by model 1 analysis and 0.20 mm/y (95% confidence interval, -0.26 to -0.14; P < .001) by model 2 analysis. A subset analysis of 7462 patients with baseline AAA size of 35 to 49 mm showed a similar inhibitory effect (1.4 ± 2.0 mm/y to 1.7 ± 2.2 mm/y; P < .001). Patients' factors associated with an increased yearly AAA growth rate were baseline AAA size, metastatic solid tumors, active smoking, chronic obstructive pulmonary disease, and chronic renal disease. Factors associated with decreased yearly AAA growth rates included prescriptions for angiotensin II type 1 receptor blockers or sulfonylureas and the presence of diabetes-related complications. Conclusions: In a nationwide analysis of diabetic Veterans Affairs patients, prescription for metformin was associated with decreased AAA enlargement. These findings provide further support for the conduct of prospective clinical trials to test the ability of metformin to limit progression of early AAA disease.