Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Feb 15.
Published in final edited form as: Circ Res. 2019 Feb 15;124(4):631–646. doi: 10.1161/CIRCRESAHA.118.312439

Compendium: Pharmacologic Management of Aneurysms

Jan H Lindeman 1, Jon S Matsumura 2
PMCID: PMC6386187  NIHMSID: NIHMS1518900  PMID: 30763216

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 (AAA) identified hundreds of candidate strategies for stabilization, and data from pre-operative clinical intervention studies show that interventions in the pathways of the activated inflammatory and proteolytic cascades in enlarging AAA are feasible. Similarly, extensive series of studies in the murine models of Marfan syndrome-related aortapathy inherited aortic root aneurysm support the concept of pharmaceutical aorta stabilization in Marfan syndrome.

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 pathologic 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.

Keywords: Aneurysm, Pharmacology, abdominal aortic aneurysm, Marfan syndrome, aneurysm, growth, pharmacologic treatment

Summary

The inconsistent correlations of preclinical successes and clinical trial results provide impetus for major advances in research innovation of aortic aneurysm. There are several explanations for this translational gap, including inadequate animal models, incomplete understanding of late stage human pathogenesis, and poorly-designed, underpowered clinical trials. Development of humanized models, advancing beyond early chemically-provoked models to those consistently demonstrating sequential dilation and rupture, addressing dysfunctional repair mechanisms, clinical studies utilizing digitized pharmaceutical 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 enlargement, run-in periods ensuring populations with active aortic enlargement prior to randomization of subjects, greater attention to concomitant medical therapy of cardiovascular risk factors, and longer follow up with clinical endpoints in clinical trials are all areas of opportunity in this important field.


An aneurysm is a localized dilatation of larger blood vessels 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 haemorrhage, 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 helpful attribution is that of primary and secondary aneurysms. Primary aneurysms relate to a matrix defect in vessel wall (i.e. fibrillin deficiency in Marfan syndrome, Collagen III deficiency in the vascular type Ehlers Danlos syndrome,4 and unknown defect(s) in aneurysms associated with bicuspid aortic valves).5,6 Secondary aneurysms relate to extensive matrix turnover and pathological vessel wall remodelling5,7 in response to a primary inflammatory insult (i.e. infection, immune diseases (Kawasaki Syndrome, Giant Cell Arthritis, Behçet syndrome),8 and the degenerative abdominal aortic aneurysm (AAA)).1

Although aneurysms occur throughout the vascular tree, there is a remarkable topographic distribution for most aneurysms (i.e. 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 embryologic origins of the vascular tree,9 which result in a persistent regional diversity in microvascular endothelium,10 mesenchymal cell characteristics11,12,13 and immunologic make-up14,15. The remarkable 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 associated with Marfan’s syndrome as clinical intervention data is available for these aneurysms. Systemic immune suppression in the context of auto-immune 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 paper.

The Abdominal Aortic Aneurysm (AAA)

An AAA is a localized dilatation caused by segmental weakening of the terminal aorta segment. The prevalence of the disease depends on the population studied, with reported prevalences varying between 1.4 and 12.4%.16 The disease carries a complex genetic predisposition17,18 and predominantly affects elderly men with a history of smoking.19

AAA’s are generally asymptomatic, and are usually diagnosed by screening or as an incidental finding. The natural history of the disease is that of slow progression and ultimate rupture.20 Ruptured AAA is a dramatic catastrophe, and aortic emergencies constitute one of the leading causes of acute death in elderly males.1,16 Risk of rupture is minimal in small aneurysms (i.e. less than 50 mm), but progressively increases with enlarging AAA size-- estimated annual rupture risks are less than 1% for AAA with a diameter of 50 mm to over 30% for an AAA exceeding 80 mm diameter.21

AAA management has been centred for decades on surgical repair of larger aneurysms to mitigate the risks of rupture. Multiple trials have shown no benefit of repair of AAA at sizes below 55 mm diameter, and consequently current guidelines advise watchful waiting for aneurysms smaller than 55 mm and preventive repair once the AAA grows over 55 mm,1,19,21 possibly with a slightly lower intervention threshold for repair in women.22

The two surgical options for repair are: open repair (through a trans-peritoneal or retroperitoneal approach) or endovascular repair (EVAR) (through a trans-arterial approach).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 patients’ preferences and characteristics such as frailty and obesity.19,21 The majority of patients is currently managed by EVAR. Open repair comes with significant higher perioperative mortality and morbidity; registry-based studies report 30-day mortality rates of approximately 4–5% for men and 6–8% 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 re-intervention, 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 that 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 --keep small aneurysms small and thereby prevent or reduce the need for surgical repair--could be advantageous. This has natural appeal to patients and from an economic point of view.27 Moreover, medical aneurysm stabilization could be beneficial as add-on strategy in patients considered at high risk for endoleak.28 It is conceivable that in patients who have aneurysm neck prone to dilation, that stabilization of the neck could reduce the incidence of late type Ia endoleak. All in all, medical 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 proteolytic imbalance.20,30 Consequently, proposed interventions directly or indirectly aim at targeting aspects of the inflammatory response, or at rectifying the proteolytic imbalance. The pertinence of these strategies is supported by a wealth of preclinical 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−/− model.31,32

The elastase model is based on a transient exposure (generally brief intra-aortic exposure) of an isolated infra-renal aorta segment with porcine pancreatic elastase.33 The rationale behind the model is the notion that loss of elastin is one of the most notable features of AAA disease. Yet, although the disease is undoubtedly characterized by extensive loss of elastin, 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 a very early phenomenon in clinical AAA development, and the elastolysis is virtually complete before the disease reaches the critical 55 mm diameter threshold.34 Secondly, clinical experience shows that chemical or surgical (endarteriectomy) does not result aneurysm formation.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 al show that the resilience of the wall essentially relies on vascular collagen.37 This phenomenon is also reflected in the dynamics of the elastase model in which exposure to pure elastin does not immediately induce AAA formation,38 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 formation is secondary and reflects a delayed, secondary response, resulting from a secondary inflammatory response.37

Although the model is referred as the ‘elastase’ model, exposure to pure pancreatic elastase does not elicit aneurysm formation.38 As such contaminants of the porcine pancreatic 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 relevance of the inflammatory response in model.

AAA formation in the elastase models follow a typical pattern with the initial moderate dilatation resulting from loss of elastic recoil, followed by a secondary dilatation, the actual aneurysm formation approximately one week after the elastase induction presumably as result of a secondary inflammatory response. The ultimately dilatation reached varies between 150–200%.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-6 neutralization.40 Other reports show that interference with the healing response either by TGFβ neutralization41 or 3-aminopropionitrile feeding induced LOX-inhibition42 elicit rupture 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 superior 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 observation 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 angiotensin model may not, or only partially translate to human AAA disease.

Based on experiments in these three models several hundred targets31 have been proposed to limit aneurysm growth. Although a detailed review of the interventions is beyond the scope of this paper, successfully targeted main clusters for intervention include: vascular inflammation, tissue remodelling, blood pressure regulation and lipid metabolism. An overview of the reported main clusters, and illustrative exemplary studies are provided in table 1.

Table 1.

Summary of successful experimental targets for pharmaceutical AAA stabilization.

Targeted cluster Strategy
Anti-inflammatory NFκB,54 AP-1,55 Rho kinase56
inhibition IL1,57 TNFα,58 CCL-159
B-cell60; γδT-cell61
depletion Neutrophil inhibition62
Mast cell inhibition63
Complement inhibition64,65
Oxilipin inhibition66,67
Immune suppression68,69
Protease inhibition MMP inhibition70,71
Cysteine protease inhibition72,73
Serine protease inhibition74,75
Oxidative stress Antioxidant enzymes76,77
Secondary antioxidants78,79
Blood pressure lowering B-blockers80
Ca-Antagonists81
ACE-inhibitors82,83
ATR-1 antagonists84
iNOS inhibition85
Lipid metabolism Statins86,87
HDL88
RXR and PPARα/γ activation89,90
Cell Therapy Mesenchymal stem cells91,92
Fibroblasts93
Matrix/Morphogens Interference with TGFβ signalling94
Interference with NOTCH95/Wnt96 signalling
Thrombospontin inhibition97
EGFR inhibition98
Metabolism Inhibition of HIF1α99
Activation of AMPK100
Nutriceuticals Polyphenols101
Phyto-oestrogens102
Sex hormones Castration103
Oestrogens104

The available literature is dominated by positive studies, few studies report disease aggravation46,47 or failure of the studied intervention.48,49,50,51 With respect to the latter, most failing interventions are included as a secondary finding presented along with a successful primary finding, raising the possibility that the available literature is biased by selective reporting of positive findings52 and type-I errors (false positive conclusions). Strong support for the latter stems from a very elegant evaluation by Trachet et al.53 The authors performed a meta-analysis of the incidence of dissecting aneurysms, and the mortality rates in the control arm of 194 papers applying the angiotensin model. The analysis indicated a strong inverse relationship between the aneurysm incidence and mortality rate of the control group, and the final conclusions of the study (median dissecting AAA incidence: 73% in studies reporting interventions claimed to reduce AAA formation, 56% in descriptive studies) vs. 35% for interventions claimed to enhance AAA formation.53 Reported median mortality rates followed the similar inverse trend: respectively: 25, 19 and 13%).53

Medical therapy for AAA patients

There are two indications for medical therapy in AAA: cardiovascular risk management and pharmaceutical AAA stabilization.

Epidemiological105,106 and cohort studies107,108,109 characterise an AAA as a strong cardiovascular risk factor. In fact, in patients deemed unfit for repair, the risk of dying from non-aneurysm-related (in particular cardiovascular) causes by far exceeds the risk of dying from the 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 indirect risk of AAA disease.111

Level IIb evidences suggests that cardiovascular risk management is effective in AAA patients,112,113 hence there is a case for cardiovascular risk management for all AAA patients, irrespective of a possible impact of the risk management on aneurysm progression. Logically, improvement in survival due to reduced cardiovascular risk not only improves the cost-effectiveness of AAA repair, but longer survival will maximize the benefits of an effective pharmaceutical stabilization program.

Preclinical models show the potential of lipid lowering,8690 antihypertensive therapy8084 and platelet aggregation inhibitors67 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 associations between β-blocker use and aneurysm progression in two small (n of 12 and 38) case control studies.114,115 These studies were then followed by a further case control116 and cohort study,117 and later by two 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% drop out rate in the treatment arm.119

The ACE-inhibitors are the second class of anti-hypertensive drugs that received significant attention in the context of AAA stabilization. Enthusiasm was spurred by supportive evidence from experimental studies,82,83 and a population-based case-control study that reported an beneficial association between ACE inhibitor use and risk of rupture).120 This study was followed by a series of non-confirmative studies,121,122,123 one of them suggesting at an adverse association between ACE-inhibitor use and AAA progression.123 These controversies were ultimately addressed in the AARDVARK study.124 This study concluded that, despite more effective blood pressure lowering, the ACE-inhibitor perindopril did not show significant impact on aneurysm growth (compared to 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 minor effect sizes. Although the authors attribute this shortcoming to the high level medical cardiac risk management in the population studied,124 it is likely that the lower than anticipated growth reflects inclusion of a disproportionate group of patients with relatively small AAAs (approx. 35 mm).

At this point the potential of the type 1 angiotensin-receptor antagonist Telmisartan is under investigation in the TEDY study.125 The rationale for this study is the fact that AT1-receptor antagonists interfere with the negative aspects of angiotensin signalling, but preserve signalling through the ATR1-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 between blood pressure and AAA progression.

The potential of statins has been evaluated in 12 studies. Results of these studies are mixed with six studies hinting at a beneficial association between statin use and AAA progression,129,130,131,132,133,134 and another six studies failing to show an association between statin use and aneurysm progression.117,122,123,135,136,137 Conclusion from the studies segregate, with the older and smaller studies being confirmative, and the later and larger studies being non-confirmative. On this basis, while the cardiovascular 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 six studies. Beneficial effects have been observed in medium sized cohort study (n=148) of patients under surveillance of a 40–49 mm AAA.138 Unfortunately, the validity of the study is challenged by the unrealistic high growth rate in the control group (5.2 mm/year; anticipated growth rate 2–3 mm/year139. A potential effect for combined aspirin-statin treatment has been observed in a sub-analysis of a study evaluating the effect of azithromycin on AAA progression.140 A benefit for NSAIDs has been reported on the basis of a very small (n=19) study reportedly patients using NSAID showed reduced AAA progression.141

In contrast, three larger studies (the UK small aneurysm trial,123 the ADAM study117 and an Australian cohort study136) all fail to confirm a beneficial effect of anti-platelet therapy on AAA progression.

Well-established negative (beneficial) associations exist between diabetic disease and AAA growth rate.142 Although this has been attributed to diabetes-related factors such as matrix stabilization by enhanced glycation and modulation of inflammation,143 there are indications that this negative (protective) association relate to off-targets effect of metformin, a biguanide antidiabetic that is first-line medication for type II diabetes. Indeed, metformin use but other classes of anti-diabetic drug associated with reduced AAA growth rate.144,145 At this point two 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 further 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 perpetuation of vascular disease including AAA at the millennium époque resulted in three studies with aimed at anti-chlamydia eradication. Two small trials with respectively a single four week course of the antibiotic roxithromycin146 or repeated (annual) four 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 sixteen weeks of macrolide treatment on AAA progression.148

A further series of clinical studies aimed at targeting specific aspects of the vascular inflammation and proteolytic imbalance in AAA. In this respect, there is a longstanding interest in the tetracycline antibiotic doxycycline. Independent of its antibiotic properties doxycycline has been shown to reduce the expression of matrix metalloproteinases,149 and to quench their activity.150 Doxycycline effectively interferes with aneurysm formation in some151,152 but not all48 models of aneurysm formation, and clinical studies showed that doxycycline treatment reduces aortic wall MMP content and improves the proteolytic imbalance through its effect on aneurysm wall protease inhibitor levels.153,154 Three studies155,156,157 evaluated the effect of doxycycline treatment on aneurysm progression, and a fourth multi-centre randomized trial (Non-Invasive Treatment of Abdominal Aortic Aneurysm Clinical Trial (N-TA^3CT) is ongoing.158 A first study small study (n=32) evaluated patients after three months of doxycycline eradication therapy with the intent of testing the persistent chlamydia infection hypothesis.155 The report claims an effect of doxycycline therapy on aneurysm progression in the 6–12, and 12–18 month follow up intervals but no effect was seen for the initial 0–6 month interval and for the overall study period. The second, open phase II study tested the safety and feasibility of six months of doxycycline therapy in AAA patients.156 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 favourably to those of historic controls not receiving treatment. In contrast, data the Pharmaceutical Aneurysm Stabilization Trial157 testing the effect of eighteen months doxycycline (100 mg/day) failed to show a benefit of doxycycline therapy on AAA progression; on the contrary, doxycycline treatment resulted in a clinically insignificant acceleration of AAA growth. Although earlier dose-finding study found dose-equivalence for low, regular and high dose (respectively 50, 100 or 300 mg/day) doxycycline on all parameters tested,159,160 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/day158 are eagerly awaited.

A potential benefit of mast cell inhibition through the potent mast-cell stabilizer pemirolast161 was tested in the AORTA trial.162 Study results show that twelve months of mast cell inhibition is safe, but the mast cell inhibition did not influence AAA progression.162

The ACZ885 (Canakinumab) for the Treatment of Abdominal Aortic Aneurysm (AAA) study163 tested the effect of IL-1β neutralization through subcutaneous Canakinumab (150 mg) once per month for twelve months. The study enrolled 65 patients and one year growth data was obtained for 20 participants in the placebo group and 23 in the Canakinumab group. AAA progression (2.5 mm/year) was similar for both groups, and the trial was terminated for reasons of futility.163

Apart from these studies on diameter changes, there is data available for surrogate endpoints (aneurysm wall inflammation). In a small study (6 cases, 10 controls) Motoki et al164 evaluated the effect of PPAR-γ agonist pioglitzone and observed a reduction in aortic wall TNFα and MMP-9 expression. The effects of the PPARα agonist fenofibrate on circulating inflammatory markers of inflammation have been studied in the Fame trial. A randomized study of 24 weeks treatment.165 No effect was observed on the circulating markers osteopontin or kallistatin. Although the authors report an absent effect on AAA growth,165 it is important to point out that the trial was not adequately powered to detect such an effect.

A highly selective suppression of aneurysm wall inflammation was observed for the selective vitamin D receptor agonist paricalcitol. It was shown that a 2–4 week pre-operative paricalcitol treatment selectively interfered with aspects of NFAT2 mediated inflammation,166 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.166 Although plasma lipids do not associate with incident AAA disease, there are weak associations between plasma LDL levels and AAA progression.167 In this light, the observed superior effects of ezetimide/simvastatin over simvastatin alone on vascular inflammation merit attention,168 yet it is unclear how these observations relate to the apparent absence of statins on AAA progression.117, 122,123,135137

The above overview of preclinical successes and clinical failures points to a major paradox of numerous preclinical successes and clinical challenges. Preclinical studies identified hundreds of successful candidate interventions, yet this enormous investment has not produced any clinical application, and no medical therapy is currently available for the stabilization of growing AAA.

More important, the apparent translational gap between preclinical and clinical studies challenges our concepts of the processes underlying late stage AAA disease pathophysiology. Undoubtedly, AAA is associated with a sustained and comprehensive inflammatory response, uncontrolled protease activity and excess matrix turn-over.5,20,154 Short-term pre-operative intervention studies in patients undergoing open repair all proved the potential of indomethacin169, statins,137,170,171,172,173,174,175,176,177 ACE-inhibitors121 and doxycycline154,159,160 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-1beta therapy163 and mast cell162 stabilization failed to influence AAA progression. Interestingly, profound immune suppression in the context of solid organ transplantation even results in accelerated AAA progression.178,179 Although these aforementioned observations do not exclude a role for inflammation and or protease activity in AAA initiation and progression, they imply involvement of additional, so far unidentified critical factors that are unresponsive to the anti-inflammatory/anti-proteolytic therapies.

One of possible key factors is failing or defective compensatory repair. In fact, interference with compensatory repair mechanisms (stem cell function) may explain the apparent disastrous effects of intense immune suppression,180 chemotherapy181 and the unexpected negative effect of doxycycline therapy157,182 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.154 Although a higher collagen cross-link content in AAA wall samples may imply more stable collagen,183 this is actually not the case due to defects at the level of collagen fibril organization. In the healthy aortic matrix the collagen fibrils are laid down in supra-molecular, intertwined network structures. As a result, forces are distributed over the wall. Loss of this network behaviour in AAA disease fundamentally impacts the mechanical stability of the wall183 and may contribute to the aortic wall weakening in the disease.

Fatty degeneration was recently identified as another potential contributor to the weakening of the aneurysm wall.184,185 Fatty degeneration is a known phenomenon in aging and chronically injured muscle,186 and thought to be a consequence of impaired repair mechanism in the context of chronic injury.187 Gene-expression studies on AAA wall specimens suggest that progressive adipocyte accumulation associated with rupture.184,188

Unfortunately, perpetual inflammatory cycle and the impaired compensatory repair that are hallmarks of 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 models189,190 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 responses 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 repair in murine models of AAA disease also explains the apparent benefit of inducing fibrotic repair in stabilizing growing AAA in murine models.191 Since the extensive fibrosis is a hallmark of human AAA disease, and that process of fibrosis results in deposition of a brittle, poor quality matrix,154,183 it is questionable whether a profibrotic strategy will stabilize human AAA.

Considering the wealth of preclinical success and failing clinical 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 findings 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.

Marfan Syndrome

Marfan syndrome is an autosomal-dominant, multisystem connective tissue disorder. The syndrome is caused by mutations in the Fbn-1 gene region located on chromosome 15, and is estimated to affect approximately 2–3/10000 individuals. Over 1000 different Fbn-1 mutations have been associated with the syndrome195 and the syndrome has extreme heterogeneous genotype-phenotype variability196 (see table 3 for the diagnostic criteria197). Ascending, and to a slighter lesser extent descending thoracic aorta aneurysms are among the primary disquieting features of the syndrome.198,199

Table 3.

Revised Ghent criteria for diagnosing Marfan syndrome

In 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 fibrillin-1 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 fibrillin-1 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 (above 20 years old) or ≥3 in those below 20 years old) AND family history of Marfan syndrome*
*In the absence of discriminating features other syndromes.
Systemic score
• Wrist AND thumb sign – 3 (Wrist OR thumb sign – 1)
• Pectus carinatum deformity – 2 (pectus excavatum or chest asymmetry – 1)
• Hind foot deformity – 2 (plain pes planus – 1)
• Pneumothorax – 2
• Dural ectasia – 2
• Protrusio acetabuli – 2
• Reduced US/LS 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, retrognathia)
• Skin striae – 1
• Myopia >3 diopters – 1
• Mitral valve prolapse (all types) – 1
Maximum total: 20 points; score ≥7 indicates systemic involvement

The Fbn-1 gene codes for fibrillin, a structural connective tissue macromolecule that has been traditionally been considered a key chaperone in elastic fiber formation. However, involvement of tissues not containing elastin indicate roles far beyond that as a scaffold of elastin formation. Indeed, defects in the Fbn-1 gene associate with impaired collagen network formation182, and fibrillin is a complex modulator of growth factor signalling and cell function.200

The aortapathy (thoracic aneurysms, dissections) is among the leading causes of premature death in Marfan patients. Although increased awareness, improved surgical techniques, and medical therapy have significantly improved prospects, Marfan syndrome still comes with significant aorta-related mortality.201,202 In this respect, strong associations have been described between the gross genotype (dominant negative (abnormal fibrillin-1 protein) or haplo-insufficient (reduced fibrillin-1 protein)) and survival; with significant better outcomes in patients with dominant negative mutations.203,204

Aneurysms in Marfan syndrome are currently managed by medical therapy (β-blockers and possibly angiotensin-II receptor type 1 antagonists (AT2 inhibitors)), and preventive surgical repair once the aneurysm size exceeds 50 mm.205 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.206 Conclusions from this study have recently been scrutinized on basis of its small size and considerable losses during follow up, and the fact that significance was only reached upon creating a composite end point.207 In addition to this single intervention study, there are additional claims from a series of observational reports.

The largest observational study is by Silverman et al.208 The authors reported outcomes for 417 patients with ‘definite’ Marfan syndrome who were under surveillance in four referral centers. Groups were created on basis of β-blockers prescription. β-blocker usage was unknown for 84 patients, as result 191 patients taking β-blocker 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 expectancies contrast with actual data in the manuscript, and with the data for other populations for the same time interval.209 Moreover, it is important to point out that the number of patients with an age over 50 in the study was very limited,208 and as a consequence 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-effects in their analysis. Although not fully clear from the text, it appears that the study covers the period between approx. 1970–1993. 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 discussion: “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”.208

A further observational study210 on an effect of β-blockers on aortapathy reports growth data for 113 juvenile Marfan patients from two centers. Different dosing schedules were used by the two centers: an intermediate dose in the first center (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’.210 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”.210 Some concerns of this study include the authors report beneficial 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 manuscript210 for a 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.210 An extreme standard deviation in the control group (69 cm (versus 22 cm in the intermediate dose group))210 implies severe skewing of the size distribution to the right in the control group, and consequently 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.211 who retrospectively evaluated the effect of β-blockers in 155 young Marfan patients 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/year (p<0.05), an effect that increased with treatment duration”.211 Although the authors rightly point out that the increase in aortic dilatation was less in the treatment arm, this difference actually reflect the larger baseline diameter in group receiving β-blockers, as the actual aortic diameters at the age of 18 were actually similar in the two 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”211 is not justified by the data in the manuscript.

Conclusions from Ladouceur et al,211 are not confirmed in a second smaller observational study in young Marfan patients.212 This study included 63 children who were monitored for over 6 years. Thirty-four patients received β-adrenergic blockade therapy (Atenolol, 0.92 mg/kg), 29 patients not receiving 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”.212 Like the other reports this retrospective analysis is prone to bias. In particular the higher percentage of patients with a family history of Marfan syndrome in the untreated group (35% vs. 69% in the treated group) may indicate that groups were not balanced with respect to the severity or phenotype.

A further small open label study213 non-randomly assigned 58 adolescent Marfan patients to β-blocker therapy (max. dose 2 mg/kg) or the ACE inhibitor Enalapril. It was concluded that ACE inhibition resulted in favorable hemodynamic changes, and a smaller increase in aortic root diameter (0.1 (1.0) vs 5.8 (5.2) mm (mean (sd)).213 Given the study design, the small sample size and absence of a control group it is difficult to draw conclusions from this study. A report from Rossi-Foulkes et al.214 compares outcomes for pre-adolescent patients on different antihypertensive therapy (β-blockers or Ca-antagonists). The authors reported that medication favorably influenced aortic growth,214 but that it did not prevent complications. In the absence of a control group, and profound baseline differences in the medicated and non-medicated group this report should be considered inconclusive.

Taken together, this overview of reports on β-adrenergic blockade in Marfan syndrome shows a paucity of studies with adequate study designs and appropriate statistical approaches.215 As a consequence, the currently available evidence does not provide a strong rationale for β-adrenergic blockade to prevent aortapathy in patients with Marfan syndrome.216,217,218 An adequate evaluation taking into account the possibility that patient responses to β-adrenergic blockade are heterogeneous and relate to the underlying genotype219,220 is missing

Observed excessive TGFβ signalling in the aortas of murine models of Marfan syndrome, and a preventive effect of interference with TGFβ signalling through neutralizing antibodies or the angiotensin II receptor antagonists in the model221 fuelled optimism for angiotensin II receptor antagonists (“Sartans”) as a preventive treatment for aortapathy in Marfan syndrome.

Supportive observations from small (respectively 28, 20 and 18 patients) open studies in young Marfan patients,222,223,224 and a small open label study on surrogate endpoints225 were followed by one smaller and four 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.226

Forteza et al. performed a larger randomized trial and randomized 5–60 year old Marfa patients to Losartan (n = 70) or atenolol (n = 70) (both dosed at 100 mg/day in individuals over 50 kg).227 The trial results show similar aortic root and ascending aorta diameters progression in the 2 arms for the 3-year follow up.227 In a French study, incorporating 303 Marfan patients aged 10 years and older, Millerton et al. assigned patients to Losartan or placebo next to their regular treatment (86% of the participants also used β-blockers).228 It was concluded that 3-year Losartan therapy did not influence the aorta parameters tested or the need for surgery.228 Unfortunately inclusion of both young and adult patients creates considerable heterogeneity both with regard to the genotype as to aortic dilation rates potentially interfering with the ability to detect suppression of growth.

Young Marfan patients were studied in a semi-blinded study by the US Marfan network. Six hundred and eight participants 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).229 Again, the 3-year follow up showed equivalence for β-adrenergic blockade or angiotensin receptor blockade. The fourth larger randomized trial is a multicentre, open-label, randomized controlled trial with blinded assessments performed in The Netherlands.230 The COMPARE trial incorporates 233 adult participants (47% female) who were randomized to either Losartan (n = 116) or no additional treatment (n = 117). The study showed mixed effects with an effect of Losartan on root dilatation rate, but no effect on the more distal aspects of the aorta. Remarkably, a planned sub-analysis performed on the available data of the COMPARE trial230 suggests that an effect of angiotensin-II receptor blockade may depend on the type of FBN-1 mutation since it was concluded that Losartan reduced only aortic root dilatation rate in haplo-insufficient patients, and not in dominant negative patients.231

According to the trial registries, there is currently one small (n=56) on-going 4-arm trial, The Oxford Marfan Trial Version which evaluates the effect of irbesartan (150–300 mg), doxycycline (100–200 mg) and a combination of both on markers of vascular dysfunction in the Marfan syndrome in patients over 13-year of age.232

With the exception of the potential beneficial effect in adult haplo-insufficient Marfan patients the clinical trials uniformly fail to show a benefit of type I angiotensin II receptor inhibition on the aortapathy in Marfan patients. Remarkably, opposite conclusions were drawn in a meta-analysis of the published prospective trials.233 Evaluation of the meta-analysis appears to have weaknesses that resulted in an overestimation of the effect size. Specifically, the planned sub-analyses performed within the COMPARE trial230 were included as separate studies, resulting in duplication of the positive data. Further, the weight distribution attributed to the studies included in the meta-analysis233 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 differences, not only with respect to aspects of the immune and inflammatory responses but also with respect to healing, as well as the significant heterogeneous character of aorta disease in Marfan’s disease.234 Most of the preclinical work is based on mice with hypomorphic FBN1 mutations: theFbn1C1039G/+ strain, with 50% of normal; and Fbn1mgR/mgR strain with 20% of normal fibrillin-1),235 but alternative models are currently being developed.236

Given the extreme genotypical and phenotypical variation in Marfan syndrome observations, from a specific murine model may only be relevant to a subset of Marfan patients. Moreover, translatability of experimental findings can be further interfered by phenotypical aspects such age (disease stage) heterogeneity is indicated by the dimorphic effects of TGFβ neutralization in an experimental model of Marfan syndrome.237 Clinical relevance of the genotypical heterogeneity as is implicated in the sub-analysis of the Compare trial that showed an exclusive benefit of Losartan in haplo-insufficient patients.231 As such a re-evaluation of the negative trials taking along the lines of dominant negative and haplo-insufficient genotypes merits consideration.

Unfortunately, such a meta-analysis may be challenging as phenotyping was only available for one third of the patients in the large Atenolol vs Losartan trial performed by the Pediatric Heart Network.229 Genotype information is available for 84% of the participants in the Sartan trial (78% established FBN1 mutation)228 and the trial by Forteza et al (82% FBN-1 mutation)227, but information on type FBN1 is missing in the publications.

Interpretation of currently available randomized trials is further challenged by the substantial phenotypical heterogeneity in the patients studied (young vs adult patients), and by loss of sensitivity by use of Z-scores rather than root size as clinical end points.238 A meta-analysis based on all individual patient data has been announced, but conclusions are awaited at the time of writing this overview.239

A further point of debate is the suggested pivotal role of TGF-β signalling in Marfan disease.221 The rationale for angiotensin II receptor type 1 (ATR1) blocker in Marfan syndrome was based on a presumed excess TGF-β signalling as the underlying cause of aortapathy in Marfan syndrome. A critical question is whether this assumption is correct as excess TGF-β activation appears a common phenomenon in aortic aneurysm disease240,241 and may actually 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 syndrome are secondary,242,243,244,245 by the fatal consequences of TGF-β neutralizing in murine models of aneurysm,41,246 and opposite contextual (disease state) effects of TGFβ on the aorta pathology in the Marfan mouse model.237

Taking into account the currently available data, there is insufficient evidence in support for either β-adrenergic blockade or ATR1 blocker for aortapathy in Marfan syndrome. The field would benefit from a meta-analysis (and sub analysis) of the available data from the Losartan trials. Such a meta-analysis has been announced,247 but conclusions are awaited at the time of writing this overview. If this is not definitive, there is need for an adequately-powered, placebo-controlled 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 effects and superior persistence,248 ATR1 antagonists might be preferable.249

Table 2.

Ongoing and planned medical intervention studies for AAA stabilization

Study acronym Intervention Read-out
Non-Invasive Treatment of Abdominal Aortic Aneurysm Clinical Trial (N-TA^3CT)158
NCT01756833
Doxycycline 100 mg bid or placebo 2-year AAA progression (CT scan), repair or rupture
TEDY125
NCT01683084
Telmisartan (AT1 receptor antagonist) 40 mg or placebo. 1-year AAA progression (US 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 inhibitor) 1-year AAA progression, surgery or repair.
FAME192
ACTRN12612001226897
Fenofibrate (PPARα agonist) 145mg or placebo 2–4 weeks prior to 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 anti-hypertensives (AT1 receptor antagonists, ACE inhibitors, or non ARB/ACE) Not specified
Metformin Therapy in Non-diabetic AAA Patients NCT03507413 Metformin (1000 mg BID) or placebo 1-year AAA progression (CT)
LIMIting AAA with MeTformin (LIMIT trial)
Not yet registered.
Metformin or placebo 2-year AAA progression (CTA)
Inositol in the MAnaGemENt of abdominal aortic aneurysm (IMAGEN)193 Inositol or placebo 1-year AAA progression (sack volume (CT))
Aortic Aneurysm Repression with Mesenchymal Stem Cells (ARREST) trial.194 1 or 3 106 cells/kg allogenic mesenchymal cells or placebo Phase I safety trial. Circulating cytokine levels and 18-FDG/PET

Acknowledgments

Research grant funding to LUMC (Dr Lindeman): Abvie, Cardoz, Eli Lilly, The Netherlands Organisation for Health Research and Development, the Dutch Heart Foundation, and the NutsOhra Fund. Research grant funding to University of Wisconsin (Dr Matsumura): Abbott, Gore, Endologix, Cook, Medtronic, and NIH.

Sources of funding: None

Abbreviations

AAA

Abdominal Aortic Aneurysm

ACE

Angiotensin Converting Enzyme

AMPK

5’ adenosine monophosphate-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

EVAR

Endovascular Aneurysm Repair

FBN1

Fibrillin-1

HDL

High Density Lipoprotein

IL-1β

Interleukin-1β

iNOS

inducible Nitric Oxide Synthetase

LDLR

Low density Lipoprotein Receptor

LOX

Lysyl Oxidase

MMP

Matrix Metalloproteinase

NFAT2

Nuclear Factor of Activated T-cells-2

NFκB

Nuclear Factor-κB

NOTCH

Notch homolog, translocation-associated

PPARα

peroxisome proliferator-activated receptors

RXR

retinoid X receptor

sd

standard deviation

TGF-β

Transforming Growth Factor-β

TNFα

Tumor Necrosis Factor-α

Footnotes

Conflicts of interest: none

Disclosures: None

References

  • 1.Tedesco MM, Dalman RL. Arterial Aneurysms In: Rutherfords Vascular Surgery, Cronenwett J and Johnston KW (eds). Saunders Elsevier; 2010: 117–130 [Google Scholar]
  • 2.Pomposelli FB, Hamdan A. Lower extremity aneurysms. In: Rutherfords Vascular Surgery, Cronenwett J and Johnston KW (eds). Saunders Elsevier; 2010: 2110–2127. [Google Scholar]
  • 3.Roche-Nagle G, Wooster D, Oreopoulos G. Popliteal vein aneurysm. Am J Surg. 2010; 199: e5–6. [DOI] [PubMed] [Google Scholar]
  • 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] [PMC free article] [PubMed] [Google Scholar]
  • 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] [PMC free article] [PubMed] [Google Scholar]
  • 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: 1583–90. [DOI] [PubMed] [Google Scholar]
  • 7.Abdul-Hussien H, Soekhoe RG, Weber E, von der Thüsen JH, Kleemann R, Mulder A, van Bockel JH, Hanemaaijer R, Lindeman JH. Collagen degradation in the abdominal aneurysm: a conspiracy of matrix metalloproteinase and cysteine collagenases. Am J Pathol. 2007; 170: 809–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Miller DV, Maleszewski JJ. The pathology of large-vessel vasculitides. Clin Exp Rheumatol. 2011; 29: S92–8. [PubMed] [Google Scholar]
  • 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] [PMC free article] [PubMed] [Google Scholar]
  • 10.Molema G Heterogeneity in endothelial responsiveness to cytokines, molecular causes, and pharmacological consequences. Semin Thromb Hemost. 2010; 36: 246–64. [DOI] [PubMed] [Google Scholar]
  • 11.Topouzis S, Majesky MW. Smooth muscle lineage diversity in the chick embryo. Two types of aortic smooth muscle cell differ in growth and receptor-mediated transcriptional responses to transforming growth factor-beta. Dev Biol. 1996; 178: 430–45. [DOI] [PubMed] [Google Scholar]
  • 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: 2271–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Leroux-Berger M, Queguiner I, Maciel TT, Ho A, Relaix F, Kempf H. Pathologic calcification of adult vascular smooth muscle cells differs on their crest or mesodermal embryonic origin. J Bone Miner Res. 2011; 26: 1543–53. [DOI] [PubMed] [Google Scholar]
  • 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 aorta: role in regional phenotypic variability and regulation of NF-κB activity. Arterioscler Thromb Vasc Biol. 2013; 33: 1248–56. [DOI] [PubMed] [Google Scholar]
  • 15.Pryshchep O, Ma-Krupa W, Younge BR, Goronzy JJ, Weyand CM. Vessel-specific Toll-like receptor profiles in human medium and large arteries. Circulation. 2008; 118: 1276–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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] [PubMed] [Google Scholar]
  • 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] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bradley DT, Badger SA, McFarland M, Hughes AE. Abdominal Aortic Aneurysm Genetic Associations: Mostly False? A Systematic Review and Meta-analysis. Eur J Vasc Endovasc Surg. 2016; 51: 64–75. [DOI] [PubMed] [Google Scholar]
  • 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 abdominal aortic aneurysms clinical practice guidelines of the European society for vascular surgery. Eur J Vasc Endovasc Surg. 2011; 41: S1–S58. [DOI] [PubMed] [Google Scholar]
  • 20.Thompson RW, Geraghty PJ, Lee JK. Abdominal aortic aneurysms: basic mechanisms and clinical implications. Curr Probl Surg. 2002; 39: 110–230. [DOI] [PubMed] [Google Scholar]
  • 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] [PubMed] [Google Scholar]
  • 22.Tomee SM, Lijftogt N, Vahl A, Hamming JF, Lindeman JHN. A registry-based rationale for discrete intervention thresholds for open and endovascular elective abdominal aortic aneurysm repair in female patients. J Vasc Surg. 2018; 67: 735–739. [DOI] [PubMed] [Google Scholar]
  • 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 [DOI] [PubMed] [Google Scholar]
  • 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] [PubMed] [Google Scholar]
  • 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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: 1704–1711. [DOI] [PubMed] [Google Scholar]
  • 27.Baxter BT, Terrin MC, Dalman RL. Medical management of small abdominal aortic aneurysms. Circulation. 2008; 117: 1883–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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 endovascular aneurysm repair. J Vasc Surg. 2015; 62: 1119–1124 [DOI] [PubMed] [Google Scholar]
  • 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] [PubMed] [Google Scholar]
  • 30.Rizas KD, Ippagunta N, Tilson MD 3rd. Immune cells and molecular mediators in the pathogenesis of the abdominal aortic aneurysm. Cardiol Rev. 2009; 17: 201–210. [DOI] [PubMed] [Google Scholar]
  • 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] [PubMed] [Google Scholar]
  • 32.Sénémaud J, Caligiuri G, Etienne H, Delbosc S, Michel JB, Coscas R. Translational Relevance and Recent Advances of Animal Models of Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol. 2017; 37: 401–410. [DOI] [PubMed] [Google Scholar]
  • 33.Azuma J, Asagami T, Dalman R, Tsao PS. Creation of murine experimental abdominal aortic aneurysms with elastase. J Vis Exp. 2009; 29 pii: 1280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.White JV, Mazzacco SL. Formation and growth of aortic aneurysms induced by adventitial elastolysis. Ann N Y Acad Sci. 1996; 800: 97–120 [DOI] [PubMed] [Google Scholar]
  • 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 elastase (PRT-201) to the venous anastomosis of arteriovenous grafts in patients with chronic kidney disease. J Vasc Access. 2014; 15: 376–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bergamini TM, Seabrook GR, Bandyk DF, Towne JB. Symptomatic recurrent carotid stenosis and aneurysmal degeneration after endarterectomy. Surgery. 1993; 113: 580–586 [PubMed] [Google Scholar]
  • 37.Dobrin PB, Banker WH, Gley WC. Elastolytic and collagenolytic studies of arteries. Implications for the mechanical properties of aneurysms. Arch Surg. 1984; 119: 405–409 [DOI] [PubMed] [Google Scholar]
  • 38.Carsten CG 3rd, 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. [DOI] [PubMed] [Google Scholar]
  • 39.Thompson RW, Curci JA, Ennis TL, Mao D, Pagano MB, Pham CT. Pathophysiology of abdominal aortic aneurysms: insights from the elastase-induced model in mice with different genetic backgrounds. Ann N Y Acad Sci. 2006; 1085: 59–73 [DOI] [PubMed] [Google Scholar]
  • 40.Kokje VBC, Gäbel G, Koole D, Northoff BH, Holdt LM, Hamming JF, Lindeman JHN. IL-6: A Janus-like factor in abdominal aortic aneurysm disease. Atherosclerosis. 2016; 251: 139–146. [DOI] [PubMed] [Google Scholar]
  • 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 Human-Like Disease in a Nondissecting Mouse Model of Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol. 2017; 37: 2171–2181. [DOI] [PubMed] [Google Scholar]
  • 42.Lu G, Su G, Davis JP, Schaheen B, Downs E, Roy RJ, Ailawadi G, Upchurch GR Jr. A novel chronic advanced stage abdominal aortic aneurysm murine model. J Vasc Surg. 2017; 66: 232–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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 abdominal aortic aneurysm model. J Vasc Surg. 2012; 56: 455–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Saraff K, Babamusta F, Cassis LA, Daugherty A. Aortic dissection precedes formation of aneurysms and atherosclerosis in angiotensin II-infused, apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2003; 23: 1621–1626. [DOI] [PubMed] [Google Scholar]
  • 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] [PubMed] [Google Scholar]
  • 46.Liu S, Xie Z, Daugherty A, Cassis LA, Pearson KJ, Gong MC, Guo Z. Mineralocorticoid receptor agonists induce mouse aortic aneurysm formation and rupture in the presence of high salt. Arterioscler Thromb Vasc Biol. 2013; 33: 1568–1579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.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 abdominal aortic aneurysms. Am J Pathol. 2012; 181: 313–321. [DOI] [PubMed] [Google Scholar]
  • 48.Xie X, Lu H, Moorleghen JJ, Howatt DA, Rateri DL, Cassis LA, Daugherty A. Doxycycline does not influence established abdominal aortic aneurysms in angiotensin II-infused mice. PLoS One. 2012; 7: e46411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Golledge J, Cullen B, Moran C, Rush C. Efficacy of simvastatin in reducing aortic dilatation in mouse models of abdominal aortic aneurysm. Cardiovasc Drugs Ther. 2010; 24: 373–378. [DOI] [PubMed] [Google Scholar]
  • 50.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] [PubMed] [Google Scholar]
  • 51.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. [DOI] [PubMed] [Google Scholar]
  • 52.Easterbrook PJ, Berlin JA, Gopalan R, Matthews DR. Publication bias in clinical research. Lancet. 1991; 337: 867–872. [DOI] [PubMed] [Google Scholar]
  • 53.Trachet B, Fraga-Silva RA, Jacquet PA, Stergiopulos N, Segers P. Incidence, severity, mortality, and confounding factors for dissecting AAA detection in angiotensin II-infused mice: a meta-analysis. Cardiovasc Res. 2015; 108: 159–70. [DOI] [PubMed] [Google Scholar]
  • 54.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] [PubMed] [Google Scholar]
  • 55.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] [PubMed] [Google Scholar]
  • 56.Wang YX, Martin-McNulty B, da Cunha V, Vincelette J, Lu X, Feng Q, Halks-Miller M, Mahmoudi M, Schroeder M, Subramanyam B, Tseng JL, Deng GD, Schirm S, Johns A, Kauser K, Dole WP, Light DR. Fasudil, a Rho-kinase inhibitor, attenuates angiotensin II-induced abdominal aortic aneurysm in apolipoprotein E-deficient mice by inhibiting apoptosis and proteolysis. Circulation. 2005; 111: 2219–2226. [DOI] [PubMed] [Google Scholar]
  • 57.Johnston WF, Salmon M, Su G, Lu G, Stone ML, Zhao Y, Owens GK, Upchurch GR Jr, Ailawadi G. Genetic and pharmacologic disruption of interleukin-1β signaling inhibits experimental aortic aneurysm formation. Arterioscler Thromb Vasc Biol. 2013; 33: 294–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.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] [PMC free article] [PubMed] [Google Scholar]
  • 59.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] [PMC free article] [PubMed] [Google Scholar]
  • 60.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 Infiltration of Immunosuppressive Cells and Is Protective of Experimental Aortic Aneurysm. Arterioscler Thromb Vasc Biol. 2016; 36: 2191–2202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.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 phosphoinositide 3-kinase/AKT signaling. J Vasc Surg. 2018; 67: 899–908 [DOI] [PubMed] [Google Scholar]
  • 62.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 abdominal aortic aneurysm formation. Circulation. 2005; 112: 232–240. [DOI] [PubMed] [Google Scholar]
  • 63.Inoue N, Muramatsu M, Jin D, Takai S, Hayashi T, Katayama H, Kitaura Y, Tamai H, Miyazaki M. Effects of chymase inhibitor on angiotensin II-induced abdominal aortic aneurysm development in apolipoprotein E-deficient mice. Atherosclerosis. 2009; 204: 359–364. [DOI] [PubMed] [Google Scholar]
  • 64.Zhou HF, Yan H, Bertram P, Hu Y, Springer LE, Thompson RW, Curci JA, Hourcade DE, Pham CT. Fibrinogen-specific antibody induces abdominal aortic aneurysm in mice through complement lectin pathway activation. Proc Natl Acad Sci U S A. 2013; 110: E4335–4344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.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 U S A. 2012; 109: E415–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.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. [DOI] [PubMed] [Google Scholar]
  • 67.Ghoshal S, Loftin CD. Cyclooxygenase-2 inhibition attenuates abdominal aortic aneurysm progression in hyperlipidemic mice. PLoS One. 2012; 7: e44369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.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 progression through inhibition of Rac1 and c-Jun-terminal-N-kinase in endothelial cells. Arterioscler Thromb Vasc Biol. 2013; 33: 2380–2388. [DOI] [PubMed] [Google Scholar]
  • 69.Yamaguchi T, Yokokawa M, Suzuki M, Higashide S, Katoh Y, Sugiyama S, Misaki T. The effect of immunosuppression on aortic dilatation in a rat aneurysm model. Surg Today. 2000; 30: 1093–1099. [DOI] [PubMed] [Google Scholar]
  • 70.Ennis T, Jin J, Bartlett S, Arif B, Grapperhaus K, Curci JA. Effect of novel limited-spectrum MMP inhibitor XL784 in abdominal aortic aneurysms. J Cardiovasc Pharmacol Ther. 2012; 17: 417–426. [DOI] [PubMed] [Google Scholar]
  • 71.Allaire E, Forough R, Clowes M, Starcher B, Clowes AW. Local overexpression of TIMP-1 prevents aortic aneurysm degeneration and rupture in a rat model. J Clin Invest. 1998; 102: 1413–1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.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 abdominal aortic aneurysm formation in apolipoprotein E-deficient mice. Cardiovasc Res. 2012; 96: 401–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.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] [PMC free article] [PubMed] [Google Scholar]
  • 74.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] [PMC free article] [PubMed] [Google Scholar]
  • 75.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] [PubMed] [Google Scholar]
  • 76.Yu Z, Morimoto K, Yu J, Bao W, Okita Y, Okada K. Endogenous superoxide dismutase activation by oral administration of riboflavin reduces abdominal aortic aneurysm formation in rats. J Vasc Surg. 2016; 64: 737–745. [DOI] [PubMed] [Google Scholar]
  • 77.Maiellaro-Rafferty K, Weiss D, Joseph G, Wan W, Gleason RL, Taylor WR. Catalase overexpression in aortic smooth muscle prevents pathological mechanical changes underlying abdominal aortic aneurysm formation. Am J Physiol Heart Circ Physiol. 2011; 301: H355–362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.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] [PubMed] [Google Scholar]
  • 79.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] [PMC free article] [PubMed] [Google Scholar]
  • 80.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. [DOI] [PubMed] [Google Scholar]
  • 81.Miao XN, Siu KL, Cai H. Nifedipine attenuation of abdominal aortic aneurysm in hypertensive and non-hypertensive mice: Mechanisms and implications. J Mol Cell Cardiol. 2015; 87: 152–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.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] [PubMed] [Google Scholar]
  • 83.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] [PubMed] [Google Scholar]
  • 84.Lida 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] [PMC free article] [PubMed] [Google Scholar]
  • 85.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] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kalyanasundaram A, Elmore JR, Manazer JR, Golden A, Franklin DP, Galt SW, Zakhary EM, Carey DJ. Simvastatin suppresses experimental aortic aneurysm expansion. J Vasc Surg. 2006; 43: 117–124. [DOI] [PubMed] [Google Scholar]
  • 87.Shiraya S, Miyake T, Aoki M, Yoshikazu F, Ohgi S, Nishimura M, Ogihara T, Morishita R. Inhibition of development of experimental aortic abdominal aneurysm in rat model by atorvastatin through inhibition of macrophage migration. Atherosclerosis. 2009; 202: 34–40. [DOI] [PubMed] [Google Scholar]
  • 88.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] [PubMed] [Google Scholar]
  • 89.Escudero P, Navarro A, Ferrando C, Furio E, Gonzalez-Navarro H, Juez M, Sanz MJ, Piqueras L. Combined treatment with bexarotene and rosuvastatin reduces angiotensin-II-induced abdominal aortic aneurysm in apoE(−/−) mice and angiogenesis. Br J Pharmacol. 2015; 172: 2946–2960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Golledge J, Cullen B, Rush C, Moran CS, Secomb E, Wood F, Daugherty A, Campbell JH, Norman PE. Peroxisome proliferator-activated receptor ligands reduce aortic dilatation in a mouse model of aortic aneurysm. Atherosclerosis. 2010; 210: 51–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Yamawaki-Ogata A, Fu X, Hashizume R, Fujimoto KL, Araki Y, Oshima H, Narita Y, Usui A. Therapeutic potential of bone marrow-derived mesenchymal stem cells in formed aortic aneurysms of a mouse model. Eur J Cardiothorac Surg. 2014; 45: e156–165. [DOI] [PubMed] [Google Scholar]
  • 92.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] [PMC free article] [PubMed] [Google Scholar]
  • 93.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 abdominal aortic aneurysm development and rupture through tissue inhibitor of metalloproteinase-1 production. Cardiovasc Res. 2017; 113: 1364–1375. [DOI] [PubMed] [Google Scholar]
  • 94.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] [PubMed] [Google Scholar]
  • 95.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] [PMC free article] [PubMed] [Google Scholar]
  • 96.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] [PubMed] [Google Scholar]
  • 97.Krishna SM, Seto SW, Jose RJ, Biros E, Moran CS, Wang Y, Clancy P, Golledge J. A peptide antagonist of thrombospondin-1 promotes abdominal aortic aneurysm progression in the angiotensin II-infused apolipoprotein-E-deficient mouse. Arterioscler Thromb Vasc Biol. 2015; 35: 389–398. [DOI] [PubMed] [Google Scholar]
  • 98.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 aortic aneurysm in a mouse model. Clin Sci (Lond). 2015; 128: 559–565. [DOI] [PubMed] [Google Scholar]
  • 99.Yang L, Shen L, Li G, Yuan H, Jin X, Wu X. Silencing of hypoxia inducible factor-1α gene attenuated angiotensin II-induced abdominal aortic aneurysm in apolipoprotein E-deficient mice. Atherosclerosis. 2016; 252: 40–49. [DOI] [PubMed] [Google Scholar]
  • 100.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 abdominal aortic aneurysms. Oncotarget. 2017; 8: 92827–92840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Wang C, Wang Y, Yu M, Chen C, Xu L, Cao Y, Qi R. Grape-seed Polyphenols Play a Protective Role in Elastase-induced Abdominal Aortic Aneurysm in Mice. Sci Rep. 2017; 7: 9402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lu G, Su G, Zhao Y, Johnston WF, Sherman NE, Rissman EF, Lau C, Ailawadi G, Upchurch GR Jr. Dietary phytoestrogens inhibit experimental aneurysm formation in male mice. J Surg Res. 2014; 188: 326–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.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] [PMC free article] [PubMed] [Google Scholar]
  • 104.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 aneurysm in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2003; 23: 1627–1632. [DOI] [PubMed] [Google Scholar]
  • 105.Freiberg MS, Arnold AM, Newman AB, Edwards MS, Kraemer KL, Kuller LH. Abdominal aortic aneurysms, increasing infrarenal aortic diameter, and risk of total mortality and incident cardiovascular disease events: 10-year follow-up data from the Cardiovascular Health Study. Circulation. 2008; 117: 1010–1017 [DOI] [PubMed] [Google Scholar]
  • 106.Forsdahl SH, Solberg S, Singh K, Jacobsen BK. Abdominal aortic aneurysms, 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] [PMC free article] [PubMed] [Google Scholar]
  • 107.Karthikesalingam A, Bahia SS, Patterson BO, Peach G, Vidal-Diez A, Ray KK, Sharma R, Hinchliffe RJ, Holt PJ, Thompson MM. The shortfall in long-term survival of patients with repaired thoracic or abdominal aortic aneurysms: retrospective case-control analysis of hospital episode statistics. Eur J Vasc Endovasc Surg. 2013; 46: 533–541. [DOI] [PubMed] [Google Scholar]
  • 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: 808–14. [DOI] [PubMed] [Google Scholar]
  • 109.Welten GM, Schouten O, Hoeks SE, Chonchol M, Vidakovic R, van Domburg RT, Bax JJ, van Sambeek MR, Poldermans D. Long-term prognosis of patients with peripheral arterial disease: a comparison in patients with coronary artery disease. J Am Coll Cardiol. 2008; 51: 1588–1596. [DOI] [PubMed] [Google Scholar]
  • 110.Parkinson F, Ferguson S, Lewis P, Williams IM, Twine CP; South East Wales Vascular Network. Rupture rates of untreated large abdominal aortic aneurysms in patients unfit for elective repair. J Vasc Surg. 2015; 61: 1606–1612. [DOI] [PubMed] [Google Scholar]
  • 111.Bulder RMA, Bastiaannet E, Hamming JF, Lindeman JH. Equal long-term survival after elective endovascular or open AAA repair: a systematic review and meta-analysis. Br J Surg. [DOI] [PubMed] [Google Scholar]
  • 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 Systematic Review and Meta-analysis. World J Surg. 2018; 42: 3443–3450. [DOI] [PubMed] [Google Scholar]
  • 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] [PubMed] [Google Scholar]
  • 114.Leach SD, Toole AL, Stern H, DeNatale RW, Tilson MD. Effect of beta-adrenergic blockade on the growth rate of abdominal aortic aneurysms. Arch Surg. 1988; 123: 606–609. [DOI] [PubMed] [Google Scholar]
  • 115.Gadowski GR, Pilcher DB, Ricci MA. Abdominal aortic aneurysm expansion rate: effect of size and beta-adrenergic blockade. J Vasc Surg. 1994; 19: 727–731. [DOI] [PubMed] [Google Scholar]
  • 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. [PubMed] [Google Scholar]
  • 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 expansion rate. JAMA Surg. 2015; 150: 44–50. [DOI] [PubMed] [Google Scholar]
  • 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. [PubMed] [Google Scholar]
  • 119.Propranolol Aneurysm Trial Investigators. Propranolol for small abdominal aortic aneurysms: results of a randomized trial. J Vasc Surg. 2002; 35: 72–79. [DOI] [PubMed] [Google Scholar]
  • 120.Hackam DG, Thiruchelvam D, Redelmeier DA. Angiotensinconverting enzyme inhibitors and aortic rupture: a populationbased case-control study. Lancet. 2006; 368: 659–665. [DOI] [PubMed] [Google Scholar]
  • 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 aneurysm. PLoS One. 2014; 9: e111952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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] [PMC free article] [PubMed] [Google Scholar]
  • 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] [PubMed] [Google Scholar]
  • 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 aneurysms: a randomized placebo-controlled trial (AARDVARK). Eur Heart J. 2016; 37: 3213–3221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Morris DR, Cunningham MA, Ahimastos AA, Kingwell BA, Pappas E, Bourke M, Reid CM, Stijnen T, Dalman RL, Aalami OO, Lindeman JH, Norman PE, Walker PJ, Fitridge R, Bourke B, Dear AE, Pinchbeck J, Jaeggi R, Golledge J. TElmisartan in the management of abDominal aortic aneurYsm (TEDY): The study protocol for a randomized controlled trial. Trials. 2015; 16: 274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Rompe F, Artuc M, Hallberg A, Alterman M, Ströder K, Thöne-Reineke C, Reichenbach A, Schacherl J, Dahlöf B, Bader M, Alenina N, Schwaninger M, Zuberbier T, Funke-Kaiser H, Schmidt C, Schunck WH, Unger T, Steckelings UM. 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] [PubMed] [Google Scholar]
  • 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 spontaneously hypertensive rats. Hypertension. 2012; 59: 291–299. [DOI] [PubMed] [Google Scholar]
  • 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: 55–61 [DOI] [PubMed] [Google Scholar]
  • 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] [PubMed] [Google Scholar]
  • 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] [PubMed] [Google Scholar]
  • 131.Schlosser FJ, Tangelder MJ, Verhagen HJ, van der Heijden GJ, Muhs BE, van der Graaf Y, Moll FL; SMART study group. Growth predictors and prognosis of small abdominal aortic aneurysms. J Vasc Surg. 2008; 47: 1127–1133. [DOI] [PubMed] [Google Scholar]
  • 132.Karlsson L, Bergqvist D, Lindback J, Parsson 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] [PubMed] [Google Scholar]
  • 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] [PMC free article] [PubMed] [Google Scholar]
  • 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] [PubMed] [Google Scholar]
  • 135.Mosorin M, Niemela 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] [PubMed] [Google Scholar]
  • 136.Ferguson CD, Clancy P, Bourke B, Walker PJ, Dear A, Buckenham T, Norman P, Golledge J. Association of statin prescription with small abdominal aortic aneurysm progression. Am Heart J. 2010; 159: 307–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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] [PMC free article] [PubMed] [Google Scholar]
  • 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: 329–34. [DOI] [PubMed] [Google Scholar]
  • 139.RESCAN Collaborators, Bown MJ, Sweeting MJ, Brown LC, Powell JT, Thompson SG. Surveillance intervals for small abdominal aortic aneurysms: a meta-analysis. JAMA. 2013; 309: 806–13. [DOI] [PubMed] [Google Scholar]
  • 140.Karlsson L, Gnarpe J, Bergqvist D, Lindback J, Parsson 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] [PubMed] [Google Scholar]
  • 141.Franklin IJ, Walton LJ, Brown L, Greenhalgh RN, Powell JT. Vascular surgical society of Great Britain and Ireland: nonsteroidal anti-inflammatory drugs to treat abdominal aortic aneurysm. Br J Surg. 1999; 86: 707. [DOI] [PubMed] [Google Scholar]
  • 142.Patel K, Zafar MA, Ziganshin BA, Elefteriades JA. Diabetes Mellitus: Is It Protective against Aneurysm? A Narrative Review. Cardiology. 2018; 141: 107–122. [DOI] [PubMed] [Google Scholar]
  • 143.Fujimura N, Xiong J, Kettler EB, Xuan H, Glover KJ, Mell MW, Xu B, Dalman RL. Metformin treatment status and abdominal aortic aneurysm disease progression. J Vasc Surg. 2016; 64: 46–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Itoga NK, Rothenberg KA, Suarez P, Ho TV, Mell MW, Xu B, Curtin CM, Dalman RL. Metformin prescription status and abdominal aortic aneurysm disease progression in the U.S. veteran population. J Vasc Surg. 2018: epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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 abdominal aortic aneurysms. Br J Surg. 2017; 104: 1486–1493. [DOI] [PubMed] [Google Scholar]
  • 146.Vammen S, Lindholt JS, Ostergaard L, Fasting H, Henneberg EW. Randomized double blind controlled trial of roxithromycin for prevention of abdominal aortic aneurysm expansion. Br J Surg.2001; 88: 1066–1072. [DOI] [PubMed] [Google Scholar]
  • 147.Hogh A, Vammen S, Ostergaard L, Joensen JB, Henneberg EW, Lindholt JS. Intermittent roxithromycin for preventing progression of small abdominal aortic aneurysms: long-term results of a small clinical trial. Vasc Endovascular Surg. 2009; 43: 452–456. [DOI] [PubMed] [Google Scholar]
  • 148.Karlsson L, Gnarpe J, Bergqvist D, Lindback J, Parsson 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] [PubMed] [Google Scholar]
  • 149.Hanemaaijer R, Visser H, Koolwijk P, Sorsa T, Salo T, Golub LM, van Hinsbergh VW. Inhibition of MMP synthesis by doxycycline and chemically modified tetracyclines (CMTs) in human endothelial cells. Adv Dent Res. 1998; 12: 114–118. [DOI] [PubMed] [Google Scholar]
  • 150.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. [DOI] [PubMed] [Google Scholar]
  • 151.Curci JA, Petrinec D, Liao S, Golub LM, Thompson RW. Pharmacologic suppression of experimental abdominal aortic aneurysms: a comparison of doxycycline and four chemically modified tetracyclines. J Vasc Surg. 1998; 28: 1082–1093. [DOI] [PubMed] [Google Scholar]
  • 152.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] [PubMed] [Google Scholar]
  • 153.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. [DOI] [PubMed] [Google Scholar]
  • 154.Lindeman JH. The pathophysiologic basis of abdominal aortic aneurysm progression: a critical appraisal. Expert Rev Cardiovasc Ther. 2015; 13: 839–851. [DOI] [PubMed] [Google Scholar]
  • 155.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 abdominal aortic aneurysms: a randomized, double blind, placebo-controlled pilot study. J Vasc Surg. 2001; 34: 606–610. [DOI] [PubMed] [Google Scholar]
  • 156.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. [DOI] [PubMed] [Google Scholar]
  • 157.Meijer CA, Stijnen T, Wasser MN, Hamming JF, van Bockel JH, Lindeman JH. Doxycycline for stabilization of abdominal aortic aneurysms: a randomized trial. Ann Intern Med. 2013; 159: 815–823. [DOI] [PubMed] [Google Scholar]
  • 158. [January 2nd, 2019]; https://clinicaltrials.gov/ct2/show/NCT01756833, accessed.
  • 159.Abdul-Hussien H, Hanemaaijer R, Verheijen JH, van Bockel JH, Geelkerken RH, Lindeman JH. Doxycycline therapy for abdominal aneurysm: Improved proteolytic balance through reduced neutrophil content. J Vasc Surg. 2009; 49: 741–749. [DOI] [PubMed] [Google Scholar]
  • 160.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 selectively depletes aortic wall neutrophils and cytotoxic T cells. Circulation. 2009; 119: 2209–2216. [DOI] [PubMed] [Google Scholar]
  • 161.Fujimiya H, Nakashima S, Miyata H, Nozawa Y. Effect of a novel antiallergic drug, pemirolast, on activation of rat peritoneal mast cells: inhibition of exocytotic response and membrane phospholipid turnover. Int Arch Allergy Appl Immunol. 1991; 96: 62–67. [DOI] [PubMed] [Google Scholar]
  • 162.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] [PubMed] [Google Scholar]
  • 163. [January 2nd, 2019]; https://clinicaltrials.gov/ct2/show/results/NCT02007252, accessed.
  • 164.Motoki T, Kurobe H, Hirata Y, Nakayama T, Kinoshita H, Rocco KA, Sogabe H, Hori T, Sata M, Kitagawa T. PPAR-γ agonist attenuates inflammation in aortic aneurysm patients. Gen Thorac Cardiovasc Surg. 2015; 63: 565–571. [DOI] [PubMed] [Google Scholar]
  • 165.Pinchbeck JL, Moxon JV, Rowbotham SE, Bourke M, Lazzaroni S, Morton SK, Matthews EO, Hendy K, Jones RE, Bourke B, Jaeggi R, Favot D, Quigley F, Jenkins JS, Reid CM, Velu R, Golledge J. Randomized Placebo-Controlled Trial Assessing the Effect of 24-Week Fenofibrate Therapy on Circulating Markers of Abdominal Aortic Aneurysm: Outcomes From the FAME −2 Trial. J Am Heart Assoc. 2018; 7: e009866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.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] [PubMed] [Google Scholar]
  • 167.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] [PubMed] [Google Scholar]
  • 168.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] [PubMed] [Google Scholar]
  • 169.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] [PubMed] [Google Scholar]
  • 170.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] [PubMed] [Google Scholar]
  • 171.Wilson WR, Evans J, Bell PR, Thompson MM. HMG-CoA reductase inhibitors (statins) decrease MMP-3 and MMP-9 concentrations in abdominal aortic aneurysms. Eur J Vasc Endovasc Surg. 2005; 30: 259–262. [DOI] [PubMed] [Google Scholar]
  • 172.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] [PubMed] [Google Scholar]
  • 173.Hurks R, Hoefer IE, Vink A, Pasterkamp G, Schoneveld A, Kerver M, de Vries JP, Tangelder MJ, Moll FL. Different effects of commonly prescribed statins on abdominal aortic aneurysm wall biology. Eur J Vasc Endovasc Surg. 2010; 39: 569–576. [DOI] [PubMed] [Google Scholar]
  • 174.Schweitzer M, Mitmaker B, Obrand D, Sheiner N, Abraham C, Dostanic S, Meilleur M, Sugahara T, Chalifour LE. Atorvastatin modulates matrix metalloproteinase expression, activity, and signaling in abdominal aortic aneurysms. Vasc Endovascular Surg. 2010; 44: 116–122. [DOI] [PubMed] [Google Scholar]
  • 175.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] [PubMed] [Google Scholar]
  • 176.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 aneurysm wall via NF-κB. Eur J Vasc Endovasc Surg. 2012; 44: 133–137. [DOI] [PubMed] [Google Scholar]
  • 177.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] [PMC free article] [PubMed] [Google Scholar]
  • 178.Lindeman JH, Rabelink TJ, van Bockel JH. Immunosuppression and the abdominal aortic aneurysm: Doctor Jekyll or Mister Hyde? Circulation. 2011; 124: e463–5. [DOI] [PubMed] [Google Scholar]
  • 179.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] [PubMed] [Google Scholar]
  • 180.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] [PubMed] [Google Scholar]
  • 181.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; 45:13–20. [DOI] [PubMed] [Google Scholar]
  • 182.De Francesco EM, Bonuccelli G, Maggiolini M, Sotgia F, Lisanti MP. Vitamin C and Doxycycline: A synthetic lethal combination therapy targeting metabolic flexibility in cancer stem cells (CSCs). Oncotarget. 2017; 8: 67269–67286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.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 U S A. 2010; 107: 862–865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.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: 1891–1900. [DOI] [PubMed] [Google Scholar]
  • 185.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 abdominal aortic aneurysm. Sci Rep. 2016; 6: 31268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Birbrair A, Zhang T, Wang ZM, Messi ML, Enikolopov GN, Mintz A, Delbono O. Role of pericytes in skeletal muscle regeneration and fat accumulation. Stem Cells Dev. 2013; 22: 2298–2314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Joe AW, Yi L, Natarajan A, Le Grand F, So L, Wang J, Rudnicki MA, Rossi FM. Muscle injury activates resident fibro/adipogenic progenitors that facilitate myogenesis. Nat Cell Biol 2010; 12: 153–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.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 Disease. J Am Heart Assoc. 2017; 6: pii: e006798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Domergue S, Jorgensen C, Noël D. Advances in Research in Animal Models of Burn-Related Hypertrophic Scarring. J Burn Care Res. 2015; 36: e259–266. [DOI] [PubMed] [Google Scholar]
  • 190.Tashiro J, Rubio GA, Limper AH, Williams K, Elliot SJ, Ninou I, Aidinis V, Tzouvelekis A, Glassberg MK. Exploring Animal Models That Resemble Idiopathic Pulmonary Fibrosis. Front Med (Lausanne). 2017; 4: 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.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 abdominal aortic aneurysm development. J Clin Invest. 2012; 122: 497–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.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] [PMC free article] [PubMed] [Google Scholar]
  • 193.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 abdominal aortic aneurysm (IMAGEN): study protocol for a randomised controlled trial. Trials. 2017; 18: 547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.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] [PubMed] [Google Scholar]
  • 195.Groth KA, Von Kodolitsch Y, Kutsche K, Gaustadnes M, Thorsen K, Andersen NH, Gravholt CH. Evaluating the quality of Marfan genotype-phenotype correlations in existing FBN1 databases. Genet Med. 2017; 19: 772–777. [DOI] [PubMed] [Google Scholar]
  • 196.Sakai LY, Keene DR, Renard M, De Backer J. FBN1: The disease-causing gene for Marfan syndrome and other genetic disorders. Gene. 2016; 591: 279–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.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: 476–85. [DOI] [PubMed] [Google Scholar]
  • 198.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: 542–52. [PubMed] [Google Scholar]
  • 199.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: 1238–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Zeyer KA, Reinhardt DP. Fibrillin-containing microfibrils are key signal relay stations for cell function. J Cell Commun Signal. 2015; 9: 309–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.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: 124–8. [DOI] [PubMed] [Google Scholar]
  • 202.Krause KJ. Marfan syndrome: literature review of mortality studies. J Insur Med. 2000; 32: 79–88. [PubMed] [Google Scholar]
  • 203.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] [PubMed] [Google Scholar]
  • 204.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] [PubMed] [Google Scholar]
  • 205.Erbel R, Aboyans V, Boileau C, Bossone E, Bartolomeo RD, Eggebrecht H, Evangelista A, Falk V, Frank H, Gaemperli O, Grabenwöger M, Haverich A, Iung B, Manolis AJ, Meijboom F, Nienaber CA, Roffi M, Rousseau H, Sechtem U, Sirnes PA, Allmen RS, Vrints CJ; 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: 2873–926. [DOI] [PubMed] [Google Scholar]
  • 206.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] [PubMed] [Google Scholar]
  • 207.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] [PMC free article] [PubMed] [Google Scholar]
  • 208.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: 157–60. [DOI] [PubMed] [Google Scholar]
  • 209.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: 124–8. [DOI] [PubMed] [Google Scholar]
  • 210.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: 629–33. [DOI] [PubMed] [Google Scholar]
  • 211.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: 406–9. [DOI] [PubMed] [Google Scholar]
  • 212.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 pediatric patients with Marfan syndrome. J Pediatr. 2007; 150: 77–82. [DOI] [PubMed] [Google Scholar]
  • 213.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: 1125–7. [DOI] [PubMed] [Google Scholar]
  • 214.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: 1364–8. [DOI] [PubMed] [Google Scholar]
  • 215.Fitzmaurice GM, Ravichandran C. A primer in longitudinal data analysis. Circulation. 2008; 118: 2005–10. [DOI] [PubMed] [Google Scholar]
  • 216.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: 300–2. [DOI] [PubMed] [Google Scholar]
  • 217.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] [PMC free article] [PubMed] [Google Scholar]
  • 218.Koo HK, Lawrence KA, Musini VM. Beta-blockers for preventing aortic dissection in Marfan syndrome. Cochrane Database Syst Rev. 2017; 11:CD011103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.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: 1057–61. [DOI] [PubMed] [Google Scholar]
  • 220.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: 60–3. [DOI] [PubMed] [Google Scholar]
  • 221.Habashi JP, Judge DP, Holm TM, Cohn RD, Loeys BL, Cooper TK, Myers L, Klein EC, Liu G, Calvi C, Podowski M, Neptune ER, Halushka MK, Bedja D, Gabrielson K, Rifkin DB, Carta L, Ramirez F, Huso DL, Dietz HC. Losartan, an AT1 antagonist, prevents aortic aneurysm in a mouse model of Marfan syndrome. Science. 2006; 312: 117–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.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: 271–6. [DOI] [PubMed] [Google Scholar]
  • 223.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: 1477–83. [DOI] [PubMed] [Google Scholar]
  • 224.Brooke BS, Habashi JP, Judge DP, Patel N, Loeys B, Dietz HC 3rd. Angiotensin II blockade and aortic-root dilation in Marfan’s syndrome. N Engl J Med. 2008; 358: 2787–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.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: 470–5. [DOI] [PubMed] [Google Scholar]
  • 226.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 randomized, double-blind clinical trial. Acta Cardiol. 2017; 72: 616–624. [DOI] [PubMed] [Google Scholar]
  • 227.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 losartan vs. atenolol for the prevention of aortic dilation in Marfan syndrome: a randomized clinical trial. Eur Heart J. 2016; 37: 978–85. [DOI] [PubMed] [Google Scholar]
  • 228.Milleron O, Arnoult F, Ropers J, Aegerter P, Detaint D, Delorme G, Attias D, Tubach F, Dupuis-Girod S, Plauchu H, Barthelet M, Sassolas F, Pangaud N, Naudion S, Thomas-Chabaneix J, Dulac Y, Edouard T, Wolf JE, Faivre L, Odent S, Basquin A, Habib G, Collignon P, Boileau C, Jondeau G. Marfan Sartan: a randomized, double-blind, placebo-controlled trial. Eur Heart J. 2015; 36: 2160–6. [DOI] [PubMed] [Google Scholar]
  • 229.Lacro RV, Dietz HC, Sleeper LA, Yetman AT, Bradley TJ, Colan SD, Pearson GD, Selamet Tierney ES, Levine JC, Atz AM, Benson DW, Braverman AC, Chen S, De Backer J, Gelb BD, Grossfeld PD, Klein GL, Lai WW, Liou A, Loeys BL, Markham LW, Olson AK, Paridon SM, Pemberton VL, Pierpont ME, Pyeritz RE, Radojewski E, Roman MJ, Sharkey AM, Stylianou MP, Wechsler SB, Young LT, Mahony L; Pediatric Heart Network Investigators. Atenolol versus losartan in children and young adults with Marfan’s syndrome. N Engl J Med. 2014; 371: 2061–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.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: 3491–500. [DOI] [PubMed] [Google Scholar]
  • 231.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 Mutation in Marfan Syndrome. Circ Cardiovasc Genet. 2015; 8: 383–8. [DOI] [PubMed] [Google Scholar]
  • 232.https://www.clinicaltrialsregister.eu/ctr-search/search?query=eudract_number:2010-023612-14 Accessed January 2nd, 2019.
  • 233.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 syndrome: a meta-analysis of prospective randomized clinical trials. Int J Cardiol. 2016; 217: 190–4. [DOI] [PubMed] [Google Scholar]
  • 234.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: 1441–4. [DOI] [PubMed] [Google Scholar]
  • 235.Cavanaugh NB, Qian L, Westergaard NM, Kutschke WJ, Born EJ, Turek JW. A Novel Murine Model of Marfan Syndrome Accelerates Aortopathy and Cardiomyopathy. Ann Thorac Surg. 2017; 104: 657–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.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 pii:dmm031542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.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 aneurysm progression in mice suggest a combinatorial therapy for Marfan syndrome. Arterioscler Thromb Vasc Biol. 2015; 35: 911–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Curtis AE, Smith TA, Ziganshin BA, Elefteriades JA. The Mystery of the Z-Score. Aorta (Stamford). 2016; 4: 124–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Pitcher A, Emberson J, Lacro RV, Sleeper LA, Stylianou M, Mahony L, Pearson GD, Groenink M, Mulder BJ, Zwinderman AH, De Backer J, De Paepe AM, Arbustini E, Erdem G, Jin XY, Flather MD, Mullen MJ, Child AH, Forteza A, Evangelista A, Chiu HH, Wu MH, Sandor G, Bhatt AB, Creager MA, Devereux RB, Loeys B, Forfar JC, Neubauer S, Watkins H, Boileau C, Jondeau G, Dietz HC, Baigent C. Design and rationale of a prospective, collaborative meta-analysis of all randomized controlled trials of angiotensin receptor antagonists in Marfan syndrome, based on individual patient data: A report from the Marfan Treatment Trialists’ Collaboration. Am Heart J. 2015; 169: 605–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.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] [PMC free article] [PubMed] [Google Scholar]
  • 241.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 aneurysms in Marfan syndrome. Neth Heart J. 2015; 23: 116–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.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: pii:e004968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Dale M, Fitzgerald MP, Liu Z, Meisinger T, Karpisek A, Purcell LN, Carson JS, Harding P, Lang H, Koutakis P, Batra R, Mietus CJ, Casale G, Pipinos I, Baxter BT, Xiong W. Premature aortic smooth muscle cell differentiation contributes to matrix dysregulation in Marfan Syndrome. PLoS One. 2017; 12: e0186603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.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] [PubMed] [Google Scholar]
  • 245.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] [PMC free article] [PubMed] [Google Scholar]
  • 246.Mallat Z, Daugherty A. AT1 receptor antagonism to reduce aortic expansion in Marfan syndrome: lost in translation or in need of different interpretation? Arterioscler Thromb Vasc Biol. 2015; 35: e10–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Pitcher A, Emberson J, Lacro RV, Sleeper LA, Stylianou M, Mahony L, Pearson GD, Groenink M, Mulder BJ, Zwinderman AH, De Backer J, De Paepe AM, Arbustini E, Erdem G, Jin XY, Flather MD, Mullen MJ, Child AH, Forteza A, Evangelista A, Chiu HH, Wu MH, Sandor G, Bhatt AB, Creager MA, Devereux RB, Loeys B, Forfar JC, Neubauer S, Watkins H, Boileau C, Jondeau G, Dietz HC, Baigent C. Design and rationale of a prospective, collaborative meta-analysis of all randomized controlled trials of angiotensin receptor antagonists in Marfan syndrome, based on individual patient data: A report from the Marfan Treatment Trialists’ Collaboration. Am Heart J. 2015; 169: 605–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Van Wijk BL, Klungel OH, Heerdink ER, de Boer A. Rate and determinants of 10-year persistence with antihypertensive drugs. J Hypertens. 2005; 23: 2101–7. [DOI] [PubMed] [Google Scholar]
  • 249.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 Marfan Syndrome. J Am Coll Cardiol. 2018; 72: 1613–1618. [DOI] [PubMed] [Google Scholar]

RESOURCES