12
Gdovascular Research CardiovascularResearch 31 (19%) 835-846 Review Mechanisms and prevention of restenosis: from experimental models to clinical practice Christophe Bauters * , Thibaud Meurice, Martial Hamon, Eug&ne McFadden, Jean-MarcLablanche, Michel E. Bertrand Service de Cardiologie B et Himodynamique, Hbpital Cardiologique, Boulevard du Professeur J. Leclercq, 59037 Lille Cedex, France Received 19 October 1995;accepted 2 February 19% Keywords: Coronary angioplasty; Restenosis; Hyperplasia; Smooth muscle cell proliferation; Platelets; Growth factors 1. Introduction Percutaneous transluminal coronary angioplasty (PTCA) was first introduced by Andreas Gruentzig in 1977 [l] as an alternative form of myocardial revascularization for patients with coronary artery disease. During the early years of its application, PTCA was limited to patients with single proximal coronary artery disease, well preservedleft ventricular function, and stable angina refractory to medi- cal treatment. Almost twenty years later, PTCA has be- come a well establishedtechnique for myocardial revascu- larization of patients with unstable angina [2], patients with an evolving myocardial infarction [3], patients with multi- vessel disease [4], and patients with depressed left ventric- ular function [5]. However, PTCA remains limited by restenosisthat occurs in 30-60% of casesdespite a suc- cessful procedure [6-g]. Assuming 500000 PTCA proce- dures per year in the United States [lo], more than 150000 patients develop restenosis every year. Although some restenosesmay be silent, most of these patients present with recurrent angina and a significant proportion will need a new revascularization procedure. Decreasing the rate of restenosis would sharply lower the long-term cost of PTCA; in the United States, a reduction of the rate of restenosis from an hypothetical 33% to 25% might save as much as $750 million annually [IO]. Numerous agents have been used to prevent restenosis, and the results of more than 40 multicenter randomized clinical trials have now been published [11,12]. Despite intensive investiga- tion in this area, no pharmacological therapy has yet been found to be useful in preventing restenosis. The purposeof this report is twofold: first, to review the available infor- mation relevant to the mechanisms of restenosis, and, second,to review the strategies currently being explored as possible approaches to the control of coronary restenosis after PTCA. 2. Mechanisms of restenosis 2.1. The healing process afrer arterial injury 2.1 .I. Neointimal hyperplasia In response to experimental arterial injury, medial smooth muscle cells @MC) shift from a contractile to a synthetic phenotype,proliferate, migrate, and produce large amounts of extracellular matrix (Fig. 1). This growth response leads to the developmentof a neointimal thicken- ing also known as neointimal hyperplasia [13] (Fig. 2). Immediately after arterial injury with a balloon catheter, multiple factors (see below) lead to the activation of SMCs. Early markers of SMC activation such as expres- sion of nuclear oncogenes are detectableas soon as 30 min after injury [ 14,151. Induction of c-fos, c-jun, and c-myc proto-oncogenes is one of the earliest transcriptional events associated with growth factor stimulation [16] and the increasedexpression of these genesis a transient response to mitogenic stimulation persisting at most for a few hours after exposure to growth factors [17]. It has been recently demonstrated that the distribution of clfos and c-jun prod- ucts after arterial injury was concentratedin smooth mus- * Corresponding author. Tel. (+33) 20.44.53.02; Fax (+ 33) 20.53.58.74. 0008-6363/96/$15.00 0 1996Elsevier Science B.V. All rights reserved PN SOOO8-6363(96>00038-7 Time for primary review 26 days. Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739 by guest on 18 February 2018

Mechanisms and prevention of restenosis: from experimental

Embed Size (px)

Citation preview

Page 1: Mechanisms and prevention of restenosis: from experimental

Gdovascular Research

Cardiovascular Research 31 (19%) 835-846

Review

Mechanisms and prevention of restenosis: from experimental models to clinical practice

Christophe Bauters * , Thibaud Meurice, Martial Hamon, Eug&ne McFadden, Jean-Marc Lablanche, Michel E. Bertrand

Service de Cardiologie B et Himodynamique, Hbpital Cardiologique, Boulevard du Professeur J. Leclercq, 59037 Lille Cedex, France

Received 19 October 1995; accepted 2 February 19%

Keywords: Coronary angioplasty; Restenosis; Hyperplasia; Smooth muscle cell proliferation; Platelets; Growth factors

1. Introduction

Percutaneous transluminal coronary angioplasty (PTCA) was first introduced by Andreas Gruentzig in 1977 [l] as an alternative form of myocardial revascularization for patients with coronary artery disease. During the early years of its application, PTCA was limited to patients with single proximal coronary artery disease, well preserved left ventricular function, and stable angina refractory to medi- cal treatment. Almost twenty years later, PTCA has be- come a well established technique for myocardial revascu- larization of patients with unstable angina [2], patients with an evolving myocardial infarction [3], patients with multi- vessel disease [4], and patients with depressed left ventric- ular function [5]. However, PTCA remains limited by restenosis that occurs in 30-60% of cases despite a suc- cessful procedure [6-g]. Assuming 500000 PTCA proce- dures per year in the United States [lo], more than 150000 patients develop restenosis every year. Although some restenoses may be silent, most of these patients present with recurrent angina and a significant proportion will need a new revascularization procedure. Decreasing the rate of restenosis would sharply lower the long-term cost of PTCA; in the United States, a reduction of the rate of restenosis from an hypothetical 33% to 25% might save as much as $750 million annually [IO]. Numerous agents have been used to prevent restenosis, and the results of more than 40 multicenter randomized clinical trials have now been published [11,12]. Despite intensive investiga- tion in this area, no pharmacological therapy has yet been

found to be useful in preventing restenosis. The purpose of this report is twofold: first, to review the available infor- mation relevant to the mechanisms of restenosis, and, second, to review the strategies currently being explored as possible approaches to the control of coronary restenosis after PTCA.

2. Mechanisms of restenosis

2.1. The healing process afrer arterial injury

2.1 .I. Neointimal hyperplasia In response to experimental arterial injury, medial

smooth muscle cells @MC) shift from a contractile to a synthetic phenotype, proliferate, migrate, and produce large amounts of extracellular matrix (Fig. 1). This growth response leads to the development of a neointimal thicken- ing also known as neointimal hyperplasia [13] (Fig. 2).

Immediately after arterial injury with a balloon catheter, multiple factors (see below) lead to the activation of SMCs. Early markers of SMC activation such as expres- sion of nuclear oncogenes are detectable as soon as 30 min after injury [ 14,151. Induction of c-fos, c-jun, and c-myc proto-oncogenes is one of the earliest transcriptional events associated with growth factor stimulation [16] and the increased expression of these genes is a transient response to mitogenic stimulation persisting at most for a few hours after exposure to growth factors [17]. It has been recently demonstrated that the distribution of clfos and c-jun prod- ucts after arterial injury was concentrated in smooth mus-

* Corresponding author. Tel. (+33) 20.44.53.02; Fax (+ 33) 20.53.58.74.

0008-6363/96/$15.00 0 1996 Elsevier Science B.V. All rights reserved PN SOOO8-6363(96>00038-7

Time for primary review 26 days.

Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739by gueston 18 February 2018

Page 2: Mechanisms and prevention of restenosis: from experimental

836 C. Bauters et al./Cardiovascular Research 31 (19%) 835-846

HOtWlOMl Growth Mechanical

F-----m ““i”A Fad0rs SMC Activation

Proliferation ; f ECM+kds

Neointimal Hyperplasia

Fig. 1. The cascade of events leading to neointimal thickening after balloon denudation. SMC = smooth muscle cell. ECM = extracellular matrix.

cle cell nuclei [15]. The corresponding oncoproteins bind to a specific DNA sequence of target genes to stimulate their transcription and are involved in the Gl phase of the cell cycle [16]. Experiments in cell culture models have shown that the use of antibodies or of antisense RNA against nuclear oncogenes prevents cell entry into the S phase and the subsequent synthesis of DNA [18,19]. It is therefore plausible that this expression of nuclear onco- genes in the hours following angioplasty is associated with the early Gl events preceding DNA synthesis in smooth muscle cells. Recently, the use of antisense oligonucleo- tides against nuclear oncogenes was shown to inhibit neointimal thickening in vivo in various animal models [20,21], suggesting that the inhibition of this early phase can significantly affect the cascade of events leading to neointimal hyperplasia a few weeks later.

Activation of SMCs is associated with a shift from a contractile to a synthetic phenotype [22-241 and leads to proliferation, migration, and synthesis of extracellular ma- trix. Proliferation of medial SMCs is evident 24 h after experimental balloon injury and continues for at least 2 weeks [25]. At least 20 to 40% of medial SMCs are activated and enter the cell cycle between 24 h and 3 days after balloon denudation [26]; these cells then migrate to the intima through breaks in the internal elastic membrane.

Many of these neointimal cells continue to proliferate for several cycles but nearly half of the migrating cells do not synthes:ize DNA [26]. Proliferation and migration should thus be considered as two distinct mechanisms leading to neointimal thickening; as discussed below, some factors may affect SMC migration but have no effect on SMC proliferation, and vice versa [27].

In animal models, the degree of intimal thickening is maximal after 3 months [28]; the additional volume that accumulates after 2 to 4 weeks reflects the adjunctive synthesis of extracellular matrix by synthetic SMCs [22- 24,291. Experimental balloon denudation is followed by a marked increase in expression of the genes that code for collagen and elastin in the arterial wall [30]. Similarly, the reexpression of embryonic forms of fibronectin occurs in the media and adventitia of rabbit arteries 24-48 h after injury; two weeks after balloon denudation, when the neointima is formed, fibronectin mRNAs as well as the fibronectin protein accumulate in the luminal layers of the neointima [31]. As pointed out by Schwartz et al. [32], cellular components constitute only about 11% of neointi- ma1 volume, and the remainder is extracellular matrix. Given the abundance of extracellular matrix in restenotic lesions, one potential “anti-restenosis” strategy would be to reduce the matrix volume surrounding each cell; a substantial reduction in neointimal volume might be ob- tained without the need to inhibit SMC migration or proliferation.

After 2 to 3 months, SMCs return to a contractile phenotype and no further significant increase in intimal thickening occurs [33].

2.1.2. Arterial remodeling Arterial remodeling is well described in de novo athero-

sclerosis. Glagov et al. [34] observed that human coronary arteries undergo adaptive enlargement in response to pro- gressive plaque expansion and maintain the lumen area until the plaque occupies 40% of the area circumscribed by

Fig. 2. Histological cross sections of rabbit iliac arteries. Left: control (non-denuded) artery. Right: iliac artery 28 days after balloon denudation. The arrow indicates the internal elastic lamina. A = adventitia; M = media; NI = neointilna.

Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739by gueston 18 February 2018

Page 3: Mechanisms and prevention of restenosis: from experimental

C. Bauters et al./ Cardiovascular Research 31 (19%) 835-846 837

the internal elastic lamina. This compensatory enlargement may thus limit the effect of plaque development on lumen narrowing.

There is increasing experimental evidence that neointi- ma1 hyperplasia is not the sole mechanism leading to lumen renarrowing after angioplasty, and that arterial re- modeling also plays a major role in this process [35-371. In the hypercholesterolemic rabbit model, Kakuta et al. [36] showed that compensatory enlargement of the vessel (increase in internal elastic lamina area) occurs in the weeks following experimental angioplasty; this process was able to accomodate nearly 60% of the neointimal formation in response to balloon injury and limit lumen narrowing. Surprisingly, restenosis was not related to neointimal formation but to a lack of compensatory en- largement or even to some degree of vessel constriction. Vascular remodeling is thus able to limit the effect of neointimal formation on chronic lumen diameter and dif- ferences in vascular remodeling, not differences in intimal formation, account for restenosis in this model. Other reports by Post et al. [35] and Lafont et al. 1371 have also demonstrated the role of vascular remodeling in other models of restenosis.

2.2. Mechanisms of restenosis in humans

One important question is whether the pathophysiologi- cal mechanisms of restenosis demonstrated in experimental models also apply to the clinical situation. Obviously the arterial wall response after coronary angioplasty in humans is less well documented than the responses in experimental models. Nevertheless, histological studies have shown that intimal thickening in the restenotic lesion contains SMCs in an abundant extracellular matrix [38-401. Immunohisto- chemical studies [41,42] have documented phenotypic modulations of SMCs after PTCA in humans; during the 2 months following PTCA, SMCs are in a synthetic pheno- type and thereafter they revert to a contractile phenotype. Various degrees of proliferation have been demonstrated in atherectomy specimens from restenotic lesions [43,44]. Migration of SMCs has never been demonstrated after PTCA in humans; it should be noted, however, that in man there is not necessarily a need for SMC migration into the intima since SMCs are already present in the athero- sclerotic plaque.

However, as in experimental models, neointimal hyper- plasia is not the sole mechanism of restenosis in humans. There is preliminary intracoronary ultrasound evidence that vascular remodeling also occurs after angioplasty in humans [45,46]. Studies by Mintz et al. [45,46] suggested that most of the late lumen loss after balloon angioplasty was due to arterial remodeling and not to intimal forma- tion. The contribution of remodeling to restenosis after nonballoon coronary angioplasty may, however, be differ- ent. Coronary stenting, for example, may eliminate any

Fig. 3. Reendothelialization after balloon denudation. Regenerated en- dothelium was identified by immunostaining with platelet-endothelial cell adhesion-l (PE~~h4-1/~-31) related antigens. Left: incomplete reen- dothelialization; Right: complete reendothelialization.

component of arterial remodeling, either enlargement or constriction (see below).

2.3. Potential regulators of the healing process

2.3.1. The endothelium The endothelium plays a fundamental role in controlling

vessel tone and SMC proliferation. After experimental angioplasty, reendothelialization of the denuded surface occurs within weeks and may be either complete [47,48] or incomplete [49-511 depending upon the animal model studied (Fig. 3); areas where the endothelial covering has rapidly regenerated have less marked intimal thickening than areas where endothelial regeneration occurs later [49,52,53]. Endothelial regeneration is probably delayed or may even be incomplete in humans. In one study, Gravanis and Roubin [54] did not find endothelial cells at the angioplasty site in patients who died within 1 month of PTCA but found substantial reendothelialization in the later specimens. Previous experimental studies support the notion that certain functions of the endothelium - includ- ing barrier regulation of permeability, thrombogenicity, and leukocyte adherence, as well as production of growth- inhibitory molecules - are critical to the prevention of luminal narrowing by neointimal thickening [55-571. Ni- tric oxide (NO) has been identified as one of the relaxant factors synthesized and released by normal endothelium 158,591. NO may theoretically interact with the process of restenosis at several levels. First, NO has an inhibitory effect on platelet adhesion and aggregation 1601; second NO has an inhibitory effect on SMC proliferation [56,61]; third, NO may exert a beneficial effect on arterial remodel- ing [62]. When L-arginine, the physiological precursor of NO, was administered to animals before endothelial de- nudation, the degree of subsequent neointimal thickening was significantly reduced compared to that observed in control animals that did not receive r.,-arginine [55,63,64].

Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739by gueston 18 February 2018

Page 4: Mechanisms and prevention of restenosis: from experimental

838 C. Baurers er al./ Cardiovascular Research 31 (1996) 835-846

2.3.2. Platelets and the thrombotic process Immediately after experimental balloon injury, endothe-

lial denudation induces platelet adhesion and aggregation resulting in release of the constituents of their alpha gran- ules .within a few minutes [7,65]. Numerous mitogenic substances, including PDGF (Platelet Derived Growth Fac- tor), are thus released at the site of injury and may be involved in the process of SMC activation [66]. Experi- ments performed in thrombocytopoenic animals have demonstrated the fundamental role of platelets in determin- ing the extent of neointimal thickening following arterial injury [67]. In a canine model of endothelial injury, the intensity of cyclic flow variations related to platelet accu- mulation was a major determinant of neointimal thickening [68]. Recent studies have shown that thrombocytopoenia inhibits migration of activated SMCs from the media to the intima but has no effect on the initial cycle of cell prolifer- ation [65]. Coagulation proteins such as thrombin may also be implicated in the response of SMCs [69]. Thrombin has mitogenic properties for SMCs [70] and has been demon- strated to induce multiple growth-related signals in SMCs including the expression of the c-fos proto-oncogene [71]. Finally, the volume of thrombus at the PTCA site may also play a role in the subsequent restenotic process. An alter- native proposal for the cellular mechanisms leading to neointimal hyperplasia has been recently advanced by Schwartz et al. [32]; this hypothesis based on the porcine coronary injury model suggests that the volume of intra- coronary thrombus at the time of PTCA may determine the subsequent volume of neointima.

In humans, the presence of thrombus at the time of PTCA can be assessed using angioscopy [72]. We recently reported the relation of angioscopic findings at the time of PTCA to subsequent restenosis [73]. Patients who had a luminal tbrombus at the PTCA site had a much higher risk of restenosis than patients without thrombus. Similarly, when PTCA is performed in patients with an unstable coronary syndrome, a situation where intraluminal throm- bus plays a major role [74], the risk of restenosis appears higher than that observed when PTCA is performed in patients with stable angina [75,76].

2.3.3. Growth factors Growth factors released at the site of injury play a

major role in the response of SMCs to balloon injury. Platelets are an important source of PDGF, but endothelial cells, macrophages, and SMCs may themselves secrete PDGF after arterial injury [22,77-791. PDGF seems to be critical for SMC migration from media to intima, whereas its absence does not limit SMC proliferation [65,80]. Using an antibody to PDGF, Ferns et al. were able to reduce neointimal SMC accumulation after experimental angio- plasty without affecting mitogenic activity [80]. Basic fi- broblast growth factor (bFGF), an in-vivo angiogenic fac- tor [Sl] with proliferative properties for both SMCs and endothelial cells through specific receptors [82,83], may

also play a role in restenosis. SMCs within the tunica media, when damaged by an oversized balloon, may re- lease bFGF due to stretch or crush injury; the liberated bFGF can then mediate the initial wave of cell division within this layer of the blood vessel. Infusion of an antibody that neutralises bFGF reduces the first cycle of SMC replication by up to 80% in the arterial media following balloon denudation but has no effect on the resulting neointimal thickening [84]. Among the other growth factors that may participate in the restenotic pro- cess are transforming growth factor p (TGFp) and insulin growth factor type 1 (IGF-1). TGFP mRNA is increased in SMCs following arterial wall injury and reaches a maxi- mum before the phase of extracellular matrix synthesis [85,86]. TGFP is known to modulate fibronectin expres- sion [87] and may be important in the control of extracellu- lar matrix synthesis [88,89]. TGFP is produced by SMCs, platelets and endothelial cells [77,90]. The main source of IGF-1 is SMCs and its mRNA expression undergoes a lo-fold increase in the weeks following balloon denuda- tion [91].

2.3.4. Hormonal factors The renin-angiotensin-aldosterone axis has been impli-

cated in the patbogenesis of restenosis [92,93]. There is evidence that angiotensin II may modulate SMC growth in vitro [94,95]; angiotensin II infusion in rats is followed by marked SMC proliferation in the intima [%I. Powell et al. have reported that inhibitors of angiotensin converting enzyme (ACE) suppress myointimal proliferation after vas- cular injury [97]. The actions of ACE inhibitors are proba- bly not solely related to an effect on angiotensin II levels but may in part be due to an effect on bradykinin metabolism [98,99] or to an effect mediated by aldosterone [lOOI. Serotonin and other vasoactive hormones (catechol- amines, vasopressin) released at the angioplasty site are also able to induce SMC proliferation [ lOl- 1041. Re- cently, considerable interest has also been focused on endothelin, a potent vasoconsttictor peptide produced by vascular endothelial cells [105]. Endothelin binds specifi- cally to human SMCs in culture and can induce nuclear oncogene expression [106,107]. Endothelin has been shown to have mitogenic activity for rat aortic SMCs [lOS] and endothelin antagonists reduce neointimal thickening after vascular injury in vivo [109,1101.

2.3.5. Mechanical factors Mechanical factors may also play a role in the degree of

restenosis in experimental models. Large areas of endothe- lial denudation without trauma to the media lead to mild neointimal thickening despite late endothelial regeneration [ 1111; in contrast, focal endothelial denudation with sub- stantial medial trauma, combined with rupture of the inter- nal elastic lamina, is associated with marked neointimal proliferation, although the endothelium regenerates within a few days [112]. These results suggest that endothelial

Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739by gueston 18 February 2018

Page 5: Mechanisms and prevention of restenosis: from experimental

C. Bauters et al./ Cardiovascular Research 31 (1996) 83.5446 839

denudation alone is not sufficient to produce major neointi- mal thickening and that direct injury of SMCs is also an important factor. Direct injury to SMCs may induce a greater neointimal response by (1) increasing the local release of growth factors by necrotic SMCs, and (2) activa- tion of intracellular pathways leading to SMC proliferation [113,114].

heparin [ 1291 and l- to 3-month treatment with subcuta- neous low molecular weight heparin [ 1301 have been shown to be inneffective. Recent clinical trials have evaluated the more powerful antithrombin hirudin; hirudin inhibits ex- perimental restenosis [131] but does not appear to be effective in humans [ 1321.

Beside acute mechanical factors, chronic mechanical factors may also play a role. Blood flow in the injured vessel may be a determinant of subsequent restenosis; experimental studies by Kohler et al. have demonstrated greater neointimal hyperplasia in the case of decreased blood flow after angioplasty [ 115,116].

3.1.3. ACE inhibitors

3. Prevention of restenosis

3.1. Discrepancies between experimental models and hu- man restenosis

A potential role for angiotensin converting enzyme (ACE) inhibitors in the limitation of neointimal prolifera- tion has been suggested, based on the demonstration that angiotensin II plays an important role in the control of SMC growth [96]. After initially positive results in experi- mental models [97], two large clinical studies (MERCA- TOR and MARCATOR), with over 2000 patients com- bined, examining the ACE inhibitor cilazapril have failed to show any significant impact on restenosis rates [133,1341.

3.1.4. Lipid-lowering agents A large number of pharmacologic trials have examined Lovastatin, which blocks the production of mevalonic

whether systematically administered pharmacologic agents acid and the synthesis of cholesterol by inhibiting the reduce the risk of angiographic restenosis [ 11,12,117]. The enzyme HMG-Co A reductase, has been shown to inhibit overwhelming majority of these clinical pharmacologic both cell proliferation in culture [ 1351 and restenosis after studies reported to date have failed to show a significant balloon angioplasty in rabbits [ 1361. In humans, however, a reduction in the incidence of restenosis in humans. These large double-blind placebo-controlled trial has recently results are in sharp contrast with the often promising been published showing, again, no benefit of the treatment results obtained in experimental models [ 1181. in the prevention of restenosis [ 1371.

3.1 .I. Platelet antagonists Platelet adhesion and activation is an important step in

vascular healing during the first days after angioplasty. Friedman et al. [67] noted suppression of the intimal proliferative response to balloon injury in severely throm- bocytopenic rabbits. Faxon et al. [119] observed a reduc- tion in the incidence and severity of recurrent stenosis in the atherosclerotic rabbit model in animals treated with sulfinpyrazone or aspirin and dipyridamole. In humans, aspirin, the prostacyclin analogue ciprostene, the serotonin antagonist ketanserin, and several thromboxane A, antago- nists have been studied [120- 1251. While antiplatelet agents have significantly reduced the risk of acute closure, there is no evidence that they may reduce restenosis. More recently, the glycoprotein receptor antibody GP IIb/IIIa has been shown to reduce clinical events 6 months after PTCA [ 1261; angiographic studies are currently being per- formed to analyze the impact of these very potent an- tiplatelet agents on restenosis.

Fish oils containing omega-3 fatty acids may decrease LDL cholesterol, increase HDL cholesterol, and alter platelet functions [138]. Several randomized clinical trials have been performed to examine if omega-3 fatty acids can reduce restenosis and have provided conflicting results [139-1421.

3.1 S. Growth factor inhibitors

3.1.2. Anticoagulants Heparin is one of the molecules that has been most

widely used in experimental models of restenosis. Both non-fractionated heparin and the newer low molecular weight derivatives have demonstrated antiproliferative ac- tivity in animal models such as the rat and the rabbit [26,127,128]. However, in humans, short-term intravenous

Several growth factor inhibitors such as trapidil and angiopeptin have been tested in the prevention of resteno- sis. Trapidil is a platelet-derived growth factor antagonist shown to be effective in experimental models of restenosis [143]. Two trials have suggested that trapidil may have an effect on restenosis, and further study of this agent is warranted [ 144,145]. Angiopeptin, a synthetic cyclic oc- tapeptide analogue of somatostatin, has been shown to reduce neointimal hyperplasia in several different animal models of angioplasty [ 146,147]. The mechanism of this effect is unknown but is thought to be related to a local inhibition of growth factors responsible for smooth muscle cell activation [146,148]. We have recently shown that, in a rabbit model of balloon denudation, pretreatment with angiopeptin is associated with a marked inhibition of c-fos and c-jun expression 30 min after injury and with a highly significant reduction in neointimal thickening 28 days after injury [ 1491. By contrast, if angiopeptin treatment is begun 1 h after injury, no effect on neointimal thickening is observed. These findings suggest that the inhibitory effect

Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739by gueston 18 February 2018

Page 6: Mechanisms and prevention of restenosis: from experimental

840 C. Bauters et al./Cardiovascular Research 31 (195%) 835-846

of angiopeptin in this model may be, at least in part, related to an inhibitory effect on early (Gl) events of the cell cycle. In humans, however, two large clinical trials, one in Europe and one in the United States, have recently been reported to show no effect of angiopeptin in the prevention of restenosis [ 150,15 11.

3.1.6. Other agents Many other agents have been evaluated as potential

inhibitors of restenosis. Calcium channel antagonists [ 152,153], corticosteroids [ 1541, and colchicine [ 1551 have failed in man in spite of encouraging results in animals. Recently, the results of the ACCORD study [ 1561 sug- gested that molsidomine, a direct NO donor, may signifi- cantly reduce the risk of angiographic restenosis; these results are in agreement with the potential beneficial ef- fects of NO donors on the restenotic process (see above). Further studies are needed with these agents to confirm this finding.

In summary, in spite of encouraging results in animal models, no systemic pharmacologic agent has been shown conclusively to produce a clinically worthwhile reduction in restenosis after PTCA. There are at least four potential explanations for these discrepancies.

A first explanation might be inter-species differences; a drug that inhibits neointimal thickening in rats or rabbits will not necessarily achieve the same effect in humans. This possibility has led to attempts to develop more repre- sentative models of restenosis, in particular the pig model [ 157,158]. Recently, however, various drugs such as an- giopeptin 11471 have been found to be effective in the pig model but not in humans [ 150,15 1 I suggesting that the pig model will not be superior to previously developed models in terms of prediction of inhibition of restenosis in hu- mans.

Secondly, many clinical trials have used low doses or inadequate duration of therapy that can also limit the ability to obtain positive results even with an effective drug. Very high doses of drugs have been necessary to inhibit restenosis in experimental models and in most ‘of the negative clinical trials the doses utilized were much lower. The inhibition of experimental restenosis with ACE inhibitors, for example, has been achieved with doses of drugs 10-100 times higher than the doses that can be used in humans [97,133,134]. This has prompted interest in the potential role of locaily delivered drug (see below). This method would allow the local release of greater quantities of drug while minimizing systemic effects.

Third, pretreatment before PTCA may also be an impor- tant issue. As already discussed above, drugs such as angiopeptin or ACE inhibitors can significantly inhibit the early (15-30 min) phase of the SMC response to injury [149,1591; this suggests that when using these agents in humans, pretreatment may increase the effectiveness of the therapy.

Finally, most of the “anti-restenosis” therapies tested so far have been directed against neointimal hyperplasia. As discussed above, neointimal hyperplasia is not the sole mechanism of restenosis after conventional balloon angio- plasty in humans and recent studies have suggested that late vessel constriction may be the major factor. Future studies will try to provide a better understanding of vessel remodeling and to identify drugs that might affect this process. In 1995, however, the use of alternative tech- niques of endovascular revascularization was increasing. Recent studies have shown that coronary stenting may significantly decrease the incidence of restenosis compared to balloon angioplasty [ 160,161]. The mechanism(s) of restenosis within coronary stents may also be different from that of restenosis after conventional balloon angio- plasty; this implies that a treatment shown to be ineffective in preventing restenosis after PTCA will not necessarily be ineffective in preventing restenosis after coronary stenting (see Section 3.3).

3.2. Local drug-delivery

3.2.1. How to deliver? In recent years, a variety of balloon catheter systems

have been designed to allow the local delivery of therapeu- tic agents at the site of arterial injury. The first devices were the double-balloon infusion catheter and the Wolinky balloon catheter [162] (a perforated balloon allowing high pressure infusion of fluids into the vessel wall). More recently, other delivery systems such as the hydrogel-coated balloon catheter [ 1631 or the dispatch catheter [ 1641 have been made available.

3.2.2. What to deliver?

3.2.2.1. Anti-proliferative drugs. Most of the antiprolifera- tive drugs that have been effective in preventing experi- mental restenosis are potential candidates for the preven- tion of restenosis using local drug-delivery systems. As stated before, this approach may allow the local release of high quantities of drug while minimizing systemic effects. However, an important limitation of this technique is the rapidity with which the infused drug leaches from the arterial wall, and the short duration of therapeutic efficacy of this approach. In experimental models, high drug levels persist for less than 48 h after drug infusion [162]. It is probable, therefore, that if this approach is to be effective, it will be necessary either to use drugs that will interfere with the early steps of the restenotic process - such as angiopeptin [149,165] - or to develop a means by which drug efflux from the arterial wall can be retarded. One potential means of achieving this would be the delivery of drug-impregnated biodegradable microspheres into the ves- sel wall to obtain a more prolonged local drug activity [166,167].

Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739by gueston 18 February 2018

Page 7: Mechanisms and prevention of restenosis: from experimental

C. Bauters et al. /Cardiovascular Research 31 (19%) 835-846 841

3.2.2.2. Gene therapy. One genetic approach to the goal of inhibiting the smooth muscle cell response to injury is the use of antisense oligonucleotides. A single-stranded DNA sequence that is complementary to a known region of a particular mRNA is synthetized. Following introduction into cells, the DNA strand binds specifically to the com- plementary nucleic acids; the double-stranded DNA-RNA hybrid is then degraded. In most of the antisense studies performed to inhibit restenosis, the target gene has been a nuclear oncogene; c-myb [20] or c-myc [21] antisense oligonucleotides delivered in a site-specific manner inhib- ited smooth muscle cell accumulation at the site of injury when inspected a few weeks after treatment. Clinical trials will soon be designed to study the effect of local delivery of antisense oligonucleotides after PTCA in humans.

Another approach is to transfect locally a gene that may inhibit the restenotic process. Experimental studies by Steg et al. [168] have demonstrated efficient gene transfer into the vascular wall using percutaneous delivery of an aden- oviral vector; they also showed that when administered with appropriate catheters the level of transfection of extravascular organs was very low. The expression of the gene was transient (a few weeks) but this may not neces- sarily be a limitation in this indication. The question of the gene to be tmnsfected remains to be answered; recently, in vivo gene transfer of nitric oxide synthase has been shown to inhibit neointima formation in injured rat carotid arteries [169].

3.2.2.3. Indirect approach. Most of the “anti-restenosis” strategies are based on a direct inhibition of the SMC response to injury. Another strategy, the indirect approach, has recently been suggested. Asahara et al. demonstrated that a single local administration of vascular endothelial growth factor (VEGF) is sufficient to facilitate endothelial repair in a rat model of balloon injury [170]; in this study, the degree of neointimal thickening at 2 weeks and 4 weeks after balloon injury was correspondingly attenuated to a statistically significant degree in arteries treated with VEGF versus controls. These results are probably related to the inhibitory effect of the endothelium on SMC growth [56,61].

3.3. Mechanical prevention

During the past several years, “new” angioplasty de- vices including atherectomy techniques, and stents have expanded the indication for angioplasty to patients with anatomy considered suboptimal for conventional PTCA. With these devices came the hypothesis that creating a more satisfactory immediate result would reduce the risk of subsequent restenosis.

There are two main mechanical atherectomy devices with sufficient clinical experience to evaluate their poten- tial to limit restenosis: rotational atherectomy (the Rotabla-

tar) and directional coronary atherectomy. Although ran- domized studies are still needed, the rates of restenosis observed with the Rotablator do not appear to be substan- tially lower than that observed after conventional PTCA [ 171,172]. It must be pointed out that adjunctive balloon angioplasty is now performed in the majority of cases after rotational atherectomy in order to achieve the largest possi- ble acute gain; this may offset the theoretical benefits of the Rotablator (i.e., preferential ablation of fibrous or calcified tissue with minimal damage to normal structures [ 1731). Directional coronary atherectomy @CA) effec- tively removes atherosclerotic tissue but also frequently constituents of the normal vascular wall [174]. DCA allows a higher acute luminal gain than conventional PTCA, the late luminal loss, however, is also higher after DCA suggesting that atherectomy may stimulate the proliferative process [ 1751. Recently, the one year follow-up of patients randomized to either DCA or F’TCA has demonstrated a significantly higher mortality in patients treated by DCA [176]; although the mechanism(s) of this unfavorable long-term effect is unknown, these results may limit the use of this technique of revascularization in humans,

The coronary stems currently under clinical evaluation are metallic devices that are implanted permanently into the vessel wall [177,178]. Recent studies have shown that implantation of an intracoronary stent in conjunction with balloon angioplasty is not only highly effective in treating acute vessel closure due to balloon-induced dissections, but may also reduce the risk of restenosis. Two large randomized trials, the Benestent study and the Stent Restenosis Study (STRESS) have been recently completed and published [160,161]. Both trials showed that at 6 months the need for repeated revascularization (the pri- mary clinical end point) was reduced with stenting, as compared with standard angioplasty. The rates of angio- graphic restenosis were also lower in patients randomized to coronary stenting than in patients treated with conven- tional balloon angioplasty (22 vs 32%, respectively in the Benestent trial, and 32 vs 428, respectively in the STRESS trial. These angiographic results were mainly related to a larger increase in the diameter of the lumen immediately after stenting. In spite of a significantly higher late loss during follow-up in the stent groups, the net result at 6 months was still better with stenting than with standard angioplasty.

The mechanism of restenosis within coronary stems is not completely understood but preliminary intravascular ultrasound studies [179] suggest that coronary stenting effectively prevents vessel constriction and that most of the late luminal loss occurring after stent implantation is due to plaque growth (probably related to neointimal hy- perplasia). This observation, taken together with the fact that the late luminal loss after coronary stenting is almost two times higher than that observed after conventional balloon angioplasty, suggests that restenosis within coro- nary stents might be much more sensitive to therapies

Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739by gueston 18 February 2018

Page 8: Mechanisms and prevention of restenosis: from experimental

842 C. Bauters et al./Cardiouascular Research 31 (19%) 835-844

designed to inhibit neointimal hyperplasia than restenosis after standard angioplasty.

Thus, the future prevention might well be the combina- tion of a mechanical device that produces the widest possible lumen and prevents vessel constriction with a pharmacologic approach to inhibit the proliferative pro- cess. Although systemic administration of “anti- restenosis” drugs has not yet been tested to prevent restenosis after coronary stenting, it is very likely that pharmacologic inhibition of neointimal hyperplasia within coronary stents will take advantage of local delivery tech- niques. In addition to local drug delivery catheters de- scribed above, the stent itself may be coated with polymers and serve as a platform for drug delivery over relatively long periods of time [ 1801.

A remaining and important question is whether com- plete inhibition of neointimal thickening is desirable. Acute coronary events are mainly the consequence of plaque rupture and thrombus formation [74]. Proliferation of SMCs and matrix production after angioplasty may prevent per- sistent instability. When angioscopy is performed a few months after PTCA of unstable plaques [181], the angio- scopic appearance is almost unvaryingly that of a stable smooth white plaque without thrombus. Some degree of neointimal thickening may thus be beneficial. If inhibition of neointimal hyperplasia becomes feasible, it will be important to determine how much should be inhibited.

4. Conclusion

In conclusion, restenosis remains an important clinical problem. The results of large pharmacologic trials have been disappointing. Restenosis which is now known to be due to both vessel remodeling and neointimal hyperplasia may be limited in the future by a combined mechanical and pharmacologic approach. The continued attractiveness of PTCA as an alternative to medical treatment and bypass surgery for patients with coronary artery disease will de- pend upon our ability to control the restenotic process.

References

[l] Gruentzig A, Senning A, Siegenthaler WE. Nonoperative dilatation of coronary artery stenosis. Percutaneous transluminal coronary angioplasty. N Engl J Med 1979;301:61-68.

[2] deFeyter PJ, Semrys PW. Percutaneous transluminal coronary an- gioplasty for unstable angina. In: Top01 El, ed. Textbook of Interventional Cardiology. Philadelphia: WB Saunders, 1994:274- 291.

[3] Grines CL, Browne KF, Marco J, et al. A comparison of immediate angioplasty with thrombolytic therapy for acute myocardial infarc- tion. N Engl J Med 1993;328:673-679.

[4] Holmes DR Jr, Berger, PB. Complex and multivessel dilation. In: Topol El, ed. Textbook of Interventional Cardiology. Philadelphia: WB Saunders, 1994231-250.

bl

161

[71

[81

[91

WI

1111

1121

[131

[141

Ml

[161

Bauters C. McFadden EP, Lablanche JM, Quandalie P, Bertrand ME. Restenosis rate after multiple percutaneous tmnsluminal coro- nary angioplasty procedures at the same site: a quantitative angio- graphic study in consecutive patients undergoing a third angio- plasty for a second mstenosis. Circulation 1993;88:%9-974. Hillegass WB, Ohman EM, Califf RM. Restenosis: the clinical issues. In: Topol, EJ, ed. Textbook of Interventional Cardiology. Philadelphia: WB Saunders, 1994415-435. Hemnan JPR, Hermans WRM, Vos J, Serruys PW. Pharmacologi- cal approaches to the prevention of restenosis following angic- plasty. The search for the holy grail? (Part I). Drugs 1993;46: 18-52. Franklin SM. Faxon DP. Pharmacologic prevention of restenosis after coronary angioplasty: review of the randomized clinical trials. Coronary Artery Dis 1993;4:232-242. Schwartz SM, deBlois D, O’Brien ERM. The intima. Soil for atherosclerosis and restenosis. Circ Res 1995;77:445-465. Bauters C, De Groote P, Adamantidis M, et al. Proto-oncogene expression in rabbit aorta after wall injury. First marker of the cellular process leading to restenosis after angioplasty? Eur Heart J 1992;13:556-559. Miano JM, Viasic N, Tota RR, Sternerman MB. Localisation of fos and jun proteins in rat aortic smooth muscle cells after vascular injury. Am J Path01 1993;142:715-724. Pardee AB. Gl events and regulation of cell proliferation. Science 1989;246:603-608.

[17] Reed JC, Alpers JD, Nowell PC, Hoover RG. Sequential expres- sion of protooncogenes during lectin-stimulated mitogenesis of normal human lymphocytes. Proc Nat1 Acad Sci USA 1986;83:3982-3986.

[18] Riabowol KT, Vosatka RJ, Ziff EB, Lamb NJ, Feramisco JR. Microinjection of fos-specific antibodies blocks DNA synthesis in fibroblast cells. Mol Cell Biol 1988;s: 1670- 1676.

[I91 Heikkila R, Schwab G, Wickstrom E, et al. A c-myc antisense oligodesoxynucleotide inhibits entry into S phase but not progress from GO to Gl. Nature 1987;328:445-449.

[20] Simons M, Edelman ER, DeKeyser JL. Langer R, Rosenberg RD. Antisense c-myb oligonucleotides inhibit arterial smooth muscle cell accumulation in vivo. Nature 1992;359:67-70.

[21] Bennett MR. Anglin S, McEwan JR, Jagoe R, Newby AC, Evan GI. Inhibition of vascular smooth muscle cell proliferation in vitro and in vivo by c-myc antisense oligodeoxynucleotides. J Clin Invest 1994;93:820-828.

[22] Thyberg J, Hedin U, SjZilund M, et al. Regulation of differentiated properties and proliferation of arterial smooth muscle cells. Arterio- sclerosis 1990;10%6-990.

[23] Schwartz SM, Campbell CR, Campbell JH. Replication of smooth muscle cells in vascular disease. Circ Res 19%,58:427-444.

[24] Rubbia L, Gabbiani G. Phenotype des cellules musculaires lisses art&ielles et athCroscl6rose. Med/Sciences 1989;53:389-395.

[25] Majesky MW, Schwartz SM. Clowes MM, Clowes AW. Heparin regulates smooth muscle S phase entry in the injured-rat carotid artery. Cite Res 1987;61:296-300.

[26] Clowes AW, Clowes MM. Kinetics of cellular proliferation after

Vogel RA. Elective supported angioplasty registry: benefit of pro- phylactic cardiopulmonary bypass support in low ejection fraction. Circulation, 1992;86(Suppl I):]-787-I-78OIAbstract). McBride W, Lange RA, Hillis LD. Restenosis after successful coronary angioplasty. Pathophysiology and prevention. N Engl J Med 1988;318:1734-1737. Liu MW, Roubm GS, King III SB. Restenosis after coronary angioplasty. Potential biologic determinants and role of intimal hyperplasia. Circulation 1989;79:1374-1387. Bauters C, Lablanche JM. McFadden EP, Leroy F, Bertrand ME. Chnical characteristics and angiographic follow-up of patients un- dergoing early or late repeat dilation for a first restenosis. J Am Co11 Cardiol 1992;20:845-848.

Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739by gueston 18 February 2018

Page 9: Mechanisms and prevention of restenosis: from experimental

C. Bauiers et al. / Cardiovascular Research 31 (1996) 835-846 843

arterial injury. II. Inhibition of smooth muscle growth by heparin. Lab Invest 1985;52:61 l-616.

[27] Casscells W. Migration of smooth muscle and endothelial cells. Critical events in restenosis. Circulation 1992;86:723-729.

[28] Clowes AW, Reidy MA, Clowes MM. Kinetics of cellular prolifer- ation after arterial injury. I. Smooth muscle growth in the absence of endothelium. Lab Invest 1983;49:327-333.

[29] Rubbia L, Gabbiani G. The cytoskeleton of arterial smooth muscle cells during development, atheromatosis and tissue culture. J Car- diovasc Pharmacol 1989;14@upp16):S9-S11.

[30] Boyd CD, Kniep AC, Pierce RA, et al. Increased elastin mRNA levels associated with surgically induced intimal injury. Comr Tiss Res 1988; 18:65-78.

[31] Bautets C, Marotte F, Hamon M, Robert V, Samuel JL, Rappaport L. Accumulation of fetal fibronectin mRNAs after balloon denuda- tion of rabbit arteries. Circulation 1995;92:904-911.

(321 Schwartz RS, Holmes DR. Top01 El. The restenosis paradigm revisited: an alternative proposal for cellular mechanisms. J Am Co11 Cardiol 1992,20:1284-1293.

[33] Clowes AW, Clowes MM, Kocher 0, Ropraz P, Chaponnier C, Gabbiani G. Arterial smooth muscle cells in vivo: relationship between actin isoform expression and mitogenesis and their modu- lation by hepatin. J Cell Biol 1988;107:1939-1945.

[34] Glagov S, Weisenberg E, Zarins CK, Stankunavicius R, Kolettis GJ. Compensatory enlargement of human atherosclerotic coronary arteries. N Engl J Med 1987;316:1371-1375.

[35] Post MJ, Borst C, Kuntz RE. The relative importance of arterial remodeling compared with intimal hypetplasia in lumen mnarrow- ing after balloon angioplasty. A study in the normal rabbit and in the hypercholesterolemic Yucatan micropig. Circulation 1994;89:2816-2821.

[36] Kakuta T, Currier JW, Haudenschild CC, Ryan TJ, Faxon DP. Differences in compensatory vessel enlargement, not mtimal forma- tion, account for restenosis after angioplasty in the hypercholes- terolemic rabbit model. Circulation 1994;89:2809-2815.

[37] Lafont A, Guzman LA, Whitlow PL, Goormastic M, Comhill JF, Chisohn GM. Restenosis after experimental angioplasty. Intimal, medial, and adventitial changes associated with constrictive remod- eling. Circ Res 1995;76:996-1002.

[38] Austin GE, Norman NB, Hollman J, Tabei S, Phillips VF. Intimal proliferation of smooth muscle cells as an explanation for recurrent coronary artery stenosis after percutaneous transluminal coronary angioplasty. J Am COB Cardiol 1985;6:369-375.

[39] Johnson DE, Hinohara T, Sehnon MR. Braden LJ, Simpson JB. Primary peripheral arterial &noses and restenoses excised by transluminal atherectomy: a histopathologic study. J Am Co11 Car- diol 1990;15:419-425.

(401 Isner JM, Keamey M, Bauters C, et al. Use of human tissue specimens obtained in vivo by directional atherectomy to study restenosis and related human vascular disorders. Trends Cardiovasc Med 1994;4:213-221.

[41] Ueda M, Becker AE, Tsukada T, Numano F, Fujimoto T. Fibrocel- lular tissue response after percutaneous transluminal coronary an- gioplasty. An histochemical analysis of the cellular composition. Circulation 1991;83: 1327-1332.

[42] Ohara T, Nanto S, Asada S, Komamura K, Wang DY. Ultrastruc- tural study of proliferating and migrating smooth muscle cells at the site of PTCA as an explanation for restenosis. Circulation 1988;78(Suppl 11):90(Abstract).

[43] O’Brien ER, Alpers CE, Stewart DK, et al. Proliferation in primary and testenotic coronary atherectomy tissue: implications for an- tiproliferative therapy. Circ Res 1993;73:223-231.

[44] Pickering JG, Weir L, Jekanowski J, Kearney MA, lsner JM. Proliferative activity in peripheral and coronary atherosclerotic plaque among patients undergoing percutaneous revascularixation. J Clin Invest 1993;Ql: 1469-1480.

[45] Mints GS, Douek PC, Banner RF, et al. Intravascular ultrasound

comparison of de novo and restenotic coronary artery lesions. J Am COB Can-ho1 1993;21:118A-118dAbstract).

[46] Kovach JA, Mirth: GS, Kent KM, et al. Serial intravascular ultra- sound studies indicates that chronic recoil is an important mecha- nism of testenosis following tramcatheter therapy. J Am Cot1 Cardiol 1993;21:484A-4840(AbstractJ.

[47] Lindner V, Reidy MA, Fmgerle J. Regrowth of arterial endothe- lium. Denudation with minimal trauma leads to complete endothe- lial cell regrowth. Lab Invest 1989;61:556-563.

[48] Weidinger FF, McLenachan JM, Cybulski Ml, et al. Persistent dysfunction of regenerated endothelium after balloon angioplasty of rabbit iliac artery. Circulation 1990,81:1667-1679.

[49] Haudenschild CC, Schwartz SM. EndotheliaI regeneration. II. Restitution of endothelial continuity. Lab Invest 1979;41:407-418.

[50] Reidy MA, Clowes AW, Schwartz SM. Endothelial regeneration. V. Inhibition of endothelial regrowth in arteries of rat and rabbit. Lab Invest 1983;49:569-575.

[51] Reidy MA, Standaert D, Schwartz SM. Inhibition of endothelial cell regrowth. Cessation of aortic endothelial cell replication after balloon catheter denudation. Arteriosclerosis 1982;2:216-220.

[52] Fishman JA, Ryan GB, Karnovsky MJ. Endothelial regeneration in the rat carotid artery and the significance of endothelial denudation in the pathogenesis of myointinud thickening. Lab Invest 1975;32:339-351.

1531 Stemerman MB, Spaet TH, Pitiick F, Cintron J, Lejnieks I, Tie11 ML. The pattern of reendotheliahzation and intimal thickening. Am J Path01 1977;87:125-142.

[54] Gravanis MB, Roubin GS. Histopathologic phenomena at the site of percutaneous transluminal coronary angioplasty: the problem of restenosis. Hum Pathol 1989;20:477-485.

[55] Hamon M, Vallet B, Bauters C, et al. Long-term oral admmistra- tion of L-arginine reduces intimal thickening and enhances neoen- dothelium-dependent acetylcholine-induced relaxation after arterial injury. Circulation 199490:1357-1362.

[56] Garg UC, Hassid A. Nitric oxide-generating vasodilatots and 8- bromo-cyclic-guanosine monophosphate inhibit mitogenesis and proliferation of cultured rat vascular smooth muscle cells. J Clin Invest 1989;83: 1774- 1777.

[57] Guo J, Milhoan KA, Tuan RS, Lefer AM. Beneficial effect of SPM-5 185, a cysteine-containing nitric oxide donor, in rat carotid artery intimal injury. Circ Res 1994;75:77-84.

(581 Furchgott RF, Zawadzki JV. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature 1980;288:373-376.

[59] Palmer RMJ, Ferrige AG, Moncada S. Nitric oxide release ac- counts for the biological activity of endothelium-derived relaxing factor. Nature 1987;327:524-526.

[60] Radomski MW, Palmer RMJ, Moncada S. Endogenous nitric oxide inhibits human platelet adhesion to vascular endothelium. Lancet 1987;2:1057-1058.

[61] Scott-Burden T, Vanhoutte PM. The endothelium as a regulator of vascular smooth muscle proliferation. Circulation 1993;87(suppl V):V-51-v-55.

[62] Langille BL, O’Donnell F. Reductions in arterial diameter pro- duced by chronic decreases in blood flow are endothelium-depen- dent. Science 1986;231:405-407.

1631 McNamara DB, Bedi B, Aurora H, et al. L-arginme inhibits balloon catheter-induced intimal hyperplasia. Biochem Biophys Res Com- mun 1993;193:291-2%.

1641 Taguchi J, Abe J, Okasaki H, Takuwa Y, Kurokawa K. L-arginine inhibits neointimal formation following balloon injury. Life Sci 1993;53:387-392.

[65] Fingerle J, Johnson R, Clowes AW, et al. Role of platelets in smooth muscle cell proliferation and migration after vascular injury in rat carotid artery. Proc Nat1 Acad Sci USA 1989;86:8412-8416.

1661 Goldberb ID, Stemerman MB. Vascular permeation of platelet factor 4 after endothelial injury. Science 1980;209:611-612.

Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739by gueston 18 February 2018

Page 10: Mechanisms and prevention of restenosis: from experimental

844 C. Bauters et al./ Cardiovascular Research 31 (1996) 835446

[67] Friedman RJ, Sternerman MB, Wenz B, et al. The effect of thrombocytopenia on experimental arteriosclerotic lesion formation in rabbits. J Clin Invest 1977;60:1191-1201.

[68] WilIerson JT, Yao SK, McNatt J, et al. Frequency and severity of cyclic flow alternations and platelet aggregation predict the severity of neointimal proliferation following experimental coronary steno- sis and endothelial injury. Proc Nat1 Acad Sci USA 1991;88:10624-10628.

[69] Fuster V, Badimon L, Badimon JJ, Chesebro JH. The pathogenesis of coronary artery disease and the acute coronary syndromes. N Engl J Med 1992326242-250.

[70] McNamara CA, Sarembock II, Gimple LW, Fenton JW, Coughlin SR, Owens GK. Thrombin stimulates proliferation of cultured rat aottic smooth muscle cells by a proteolytically activated receptor. J Clin Invest 1993;91:94-98.

[71] Berk BC, Taubman MB, Griendling KK, et al. Thrombin stimu- lated events in cultured vascular smooth muscle cells. J Biol Chem 1990;265:1734-1740.

[72] den He&r P, Foley DP, Escaned J, et al. Angioscopic versus angiographic detection of intimal dissection and intracoronary thrombus. J Am Co11 Catdiol 1994;24:649-654.

[73] Bauters C, Labianche JM. McFadden EP, Hamon M, Bertrand ME. Relation of coronary angioscopic fmdings at coronary angioplasty to angiographic restenosis. Circulation 1995;92:2473-2479.

[74] Falk E. Unstable angina with fatal outcome: dynamic coronary thrombosis leading to infarction and or sudden death. Circulation 1985;71:699-708.

[75] De Groote P, Bauters C, McFadden EP, Lablanche JM, Leroy F, Bertrand ME. Local lesion-related factors and restenosis after coronary angioplasty: evidence from a quantitative angiographic study in patients with unstable angina undergoing double-vessel angioplasty. Circulation 1995:91:%8-972.

[76] Bauters C, Khanoyan P, McFadden EP, Quandalle P, Lablanche JM, Bertrand ME. Restenosis after delayed coronary angioplasty of the culprit vessel in patients with a recent myocatdial infarction treated by tbrombolysis. Circulation 1995;91:1410-1418.

[77] Assoian RK, Grotendorst GR, Miller DM, Spom MB. Cellular transformation by coordinate action of three peptide growth factors from human platelets. Nature 1984;309:804-806.

[78] Walker LN, Bowen-Pope DF. Ross R, Reidy MA. Production of platelet-derived growth factor-like molecules by cultured arterial smooth muscle cells accompanies proloferation after arterial injury. Prnc Natl Acad Sci USA 1986;83:731 l-7315.

[79] Wilcox JN, Smith KM, Williams LT. Schwartz SM, Gordon D. Platelet-derived growth factor mRNA detection in human athero- sclerotic plaques by in situ hybridization. J Clin Invest 1988;82: 1134- 1143.

[80] Ferns GA, Raines EW, Spmgel KH, Motani AS, Reidy MA, Ross R. Inhibition of neointimal smooth muscle accumulation after angioplasty by an antibody to PDGF. Science 199 I ;253: 1129- 1132.

[81] Lindner V, Lappi DA, Baird A, Majack RA. Reidy MA. Role of basic fibroblast growth factor in vascular lesion formation. Circ Res 1991;68:106-113.

[82] Lindner V, Majack RA, Reidy MA. Basic fibroblast growth factor stimulates endothelial regrowth and proliferation in denuded arter- ies. J Clin Invest 1990;85:200&2008.

[83] Lindner V, Majack RA, Reidy MA. Basic FGF induces the prolif- eration of vascular cells in injured arteries. FASEB J 1990;4: A625.

[84] Lindner V, Reidy MA. Proliferation of smooth muscle cells after vascular injury is inhibited by an antibody against basic fibroblast growth factor. Proc Natl Acad Sci USA 1991;88:3739-3743.

[85] Bassols A, Massaque J. Transfotming growth factor b regulates the expression and stmcture of extracellular matrix chondroitin/der- matan sulfate proteoglycans. J Biol Chem 1988;263:3039-3045.

[86] Nikol S, Isner JM, Pickering JG, Keamey M, Lecletc G, Weir L. Expression of transforming growth factor-b1 is increased in human vascular mstenosis lesions. J Clin Invest 1992.90: 1582-1592.

[87] Border WA, Ruosslshti E. Transforming growth factor-b in dis- ease: the dark side of tissue repair. J Clin Invest 1992,90: l-7.

[88] Spom MB, Roberts AB, Wakefield LM, de Combrugghe B. Some recent advances in the chemistry and biology of transforming growth factor-b. J Cell Biol 1987;105:1039-1045.

1891 Roberts AB, Spom MB, Assoiau RK, et al. Transforming growth factor type b. Rapid induction of fibrosis and angiogenesis in vivo and stimulation of collagen formation in vitro. Proc Natl Acad Sci USA 1986;83:4167-4170.

[%I Antonelli-Grlidge A, Saunders KB, Smith SR, D’Amore PA. An activated form of transforming growth factor-b is produced by cccultures of endothelial cells and pericytes. Pmc Natl Acad Sci USA 1989;86:4544-4548

[91] Cetcek B, Fishbern MC: Forrester JS, Helfant RH, Fagin JA. Induction of insulin-like growth factor I messenger RNA in rat aorta after balloon denudation. Cim Res 1990;66:1755-1760.

1921 D;r,au VJ. Vascular mnin angiotensin: a possible autocrine or paracrine system in control of vascular function. J Cardiovasc Pharmacol 1984;6:S377-S382.

[93] Rakugi H, Kim DK. Krieger JE, Wang DS, Dzau VJ, Pratt RE. Induction of angiotensiu converting enzyme in the neointima after vascular injury. Possible role in restenosis. J Clin Invest 1994;93:339-346.

1941 Berk BC, Vekhstein V, Gordon HM, Tsuda T. Angiotensin II- stimulated protein synthesis in cultured vascular smooth mucle cells. Hypertension 1989; 13:305-3 14.

1951 Turla MB, Thompson MM, Corjay MH, Owens GK. Mechanisms of angiotensin II- and arginme vasopmssin-induced increases in protein synthesis and content in cultured rat aortic smooth muscle cells. Cite Res 1991;68:288-299.

[%I Daemen MJAP, Lombardi DM, Bosman FT. Schwartz SM. An- giotensin II induces smooth muscle cell proliferation in tbe normal and injured rat arterial wall. Circ Res 1991;68:450-456.

[97] Powell JS, Miiller RKM, Kuhn H, Hefti F, Baumgartner HR. Inhibitors of angiotensin-converting enzyme prevent myointimal proliferation after vascular injury. Science 1989245: 186-188.

[98] Van Belle E. Vallet B, Auffray JL. et al. Chronic inhibition of nitric oxide synthesis reverses the effects of ACE inhibition on neo-endotbelium-dependent relaxation and on intimal thickening after balloon injury. Am J Physiol 19%,27@H298-H305.

(991 Farhy RD, Carretero OA, Ho KL, Scicli AG. Role of kinins and nitric oxide in the effects of angiotensin converting enzyme in- hibitors on neointima formation. Circ Res 1993;72:1202-1210.

[lo01 Van Belle E, Bautem C, Wemert N, et al. Neointimal thickening after balloon denudation is enhanced by aldosterone and inhibited by spironolactone, an aldosterone antagonist. Cardiovasc Res 1995;29:27-32.

[lOI] Vanhoutte PM, Houston D. Platelets, endothelium and vasospasm. Circulation 1985;72:728-734.

[ 1021 Ncmecek GM, Coughlin SR, Handley DA, Moskowitz MA. Stimu- lation of aortic smooth muscle cell mitogenesis by serotonin. Proc NaU Acad Sci USA 1986;83:674-678.

(1031 Moalic JM. Bauters C, Himbert D, et al. Phenylephrine, vase- pressin and angiotensin II as determinants of prom-oncogene and he.at-shock protein gene expression in adult rat heart and aorta. J Hypertension 1989;7: 195-201.

11041 Iwaki K. Sukhatme VP, Shubeita HE, Chien KR. Alpha- and beta-adrenergic stimulation induces distinct patterns of immediate early gene expression in neonatal rat myocardial cells. J Biol Chem 1990;265:13809-13817.

[105] Yanagisawa M, Kurihara H, Kiiura S, et al. A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature 1988;332:41 l-415.

[106] Clozel M, Fischli W. Guilly C. Specific binding of endothelin on human vascular smooth muscle cells in culture. J Clm Invest 1989;83:1758-1761.

[107] Komuro I, Kmihara H, Sugiyama T, Takaku F, Yasaki Y. Endothe-

Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739by gueston 18 February 2018

Page 11: Mechanisms and prevention of restenosis: from experimental

C. Bauters et al./ Cardiovascular Research 31 (19%) 835-846 845

lin stimulates c-fos and c-myc expression and proliferation of vascular smooth muscle cells. FEBS Lett 1988;238:249-259.

[ 1081 Hirata Y, Takagi Y, Fukuda Y, Marumo F. Endothelin is a potent mitogen for rat vascular smooth muscle cells. Arteriosclerosis 1989;78:225-228.

[IO91 Douglas SA, Louden C, Vickery-Clark LM. et al. A role for endogenous endotherm-1 in neointimal formation after rat carotid artery balloon angioplasty. Protective effects of the novel nonpep- tide endotherm receptor antagonist SB 20%70. Circ Res 1994;75:190-197.

[ 1 IO] Azuma H, Hamasaki H, Niimi Y, Terada T, Matsubara 0. Role of endothelin-1 in neointima formation after endothelial removal in rabbit carotid arteries. Am J Physiol 1994;267:H2259-H2267.

[l 1 I] Fmgerle J, Tina AUYP, Clowes AW, Reidy MA. httimal lesion formation in rat carotid arteries after endothelial denudation in absence of medial injury. Arteriosclerosis 1990; IO: lO82- 1087.

[I 121 Walker LN, Ramsay MM, Bowyer DE. Endothelial healing follow- ing defined injury to rabbit aorta: depth of injury and mode of repair. Arteriosclerosis 1983;47: l23- 130.

[II31 Kulik TJ, Rothman A. Glennon ET, Underwood RH. Stretching vascular smooth muscle causes inositol phosphates turnover. Circu- lation 1989;8dSuppl I I): 198(Abstract).

[I 141 Clowes AW, Clowes MM, Reidy MA. Role of acute distension in the induction of smooth muscle proliferation after endothelial denudation. Fed Prcc 1987;46:720.

[I 151 Kohler TR, Jawien A. Flow affects development of intimal hyper- plasia after arterial injury in rats. Arterioscler Thromb 1992;12:%3-971.

[116] Kohler TR, Kirkman TR, Kraiss LW, Zierler BK, Clowes AW. Increased blood flow inhibits neointimal hyperplasia in endothelial- ized vascular grafts. Circ Res 1991;69:1557-1565.

[I 171 Popma JJ, Califf RM, Topol EJ. Clinical trials of restenosis after coronary angioplasty. Circulation 1991;84: 1426-1436.

[118] Bauters C, Hamon M, Van Belle E. Dupuis B, Lablanche JM, Bertrand ME. Rest&rose apt& angioplastie coronaire. Apport des modeles exp&imentaux. Arch Mal Coeur 1993;86:47-56.

[ 1191 Faxon DP, Sanbom TA, Haudenschild CC, Ryan TJ. Effect of antiplatelet therapy on restenosis after experimental angioplasty. Am J Cardiol 1984$3:72C-76C.

[ 1201 Schwartz L, Bourassa MG, Lespersnce J, et al. Aspirin and dipyri- damole in the prevention of restenosis after percutaneous translumi- nal coronary angioplasty. N Engl J Med 1988;318:1714-1719.

[I211 Darius H, Nixdorff U, Zander J, et al. Effects of ciprostene on restenosis rate and platelet activation during therapeutic PTCA. Eur Heart J 199 I ; 12(Suppl):26(Abstract).

[I221 Serruys PW, Klein W, Rutsch W, et al. PARK: the Post Angio- plasty Restenosis Ketanserin Trial. J Am Coll Cardiol 1992;2 1:322A(Abstract).

[I231 Serrnys PW, Rutsch W, Heyndrickx Cl, et al. Prevention of resteno- sis after percutaneous transluminal coronary angioplasty with trom- boxan A2 receptor blockade. Circulation 1991;84: I568- 1580.

11241 Yabe Y, Okamoto K, Gosawa H, et al. Does a tbromboxane A2 syntbetase inhibitor prevent restenosis after PTCA? Circulation 1989;8O(Suppl 2):11-26OlAbstract).

[125] Finci L, Meier B, Steffenino G, Rutishauser W. Aspirin versus placebo after coronary angioplasty for prevention of restenosis. Eur Heart J 1988;9(Suppl): 156(Abstract).

[I261 Topol EJ, CaIiff RM, Weisman HF, et al. Random&d trial of coronary intervention with antibody against platelet IIb/lIIa inte- grin for reduction of clinical restenosis: results at six months. Lancet 1994;343:881-886.

[ 1271 Pukac LA, Hirsch GM, Lormeau JC, Petitou M, Choay J, Kamovsky MJ. Antiproliferative effects of novel, non anticoagulant heparin derivatives on vascular smooth muscle cells in vitro and in vivo. Am J Pathol 1991;139:1501-1509.

[I281 Hamon M, Bauters C, Wemert N, et al. Heparin does not inhibit

oncogene induction in rabbit aorta following balloon denudation. Cardiovasc Res 1993;27:1209-1213.

[I291 Ellis SG, Roubin GS, Wilentx J, Douglas JS, King SB. Effect of 18-24 hour heparin administration for prevention of mstenosis after uncomplicated coronary angioplasty. Am Heart J 1989;117:777-782.

[I301 Faxon DP, Sprio T, Minor S, et al. Enoxaparin, a low molecular weight heparin, in the prevention of testenosis after angioplasty: results of a double blind randomised trial. J Am Coll Cardiol 1992;19:258A(Abstract).

[I311 Sarembock IJ, Gertz SD, Gimple LW, et al. Effectiveness of recombinant desulphato hirudin in reducting restenosis after bal- loon angioplasty of atherosclerotic femoral arteries in rabbits. Circulation 1991;84:232-243.

[I321 Serruys PW, Deckers JW, Close P, on behalf of the HELVETICA study group. A double blind, radomized, heparin controlled-trial evaluating acute and long-term efficacy of r-hirudin (CGP 39 393) in patients undergoing coronary angioplasty. Circulation 1994;9OI- 394(Abstract).

[I331 MERCATOR study group. Does the new angiotensin converting enzyme inhibitor cilazapril prevent rcstenosis after percutaneous transluminal coronary angioplasty? Results of the MERCATOR Study: a multicenter, randomised, double-blind placebo-controlled trial. Circulation 1992;86:100-1 IO.

]I341 MARCATOR study group. Effect of high dose angiotensin-con- vetting enzyme inhibition on restenosis: final results of the MAR- CATOR study, a multicenter, double-blind, placebo-controlled trial of cilazapril. J Am Coll Cardiol 1995;2:362-369.

[I351 Constantinescu DE, Banka VS. Tulenko TN. Lovastatin inhibits proliferation of arterial smooth muscle and endothelial cells. Indica- tion in atherosclerosis and prevention of restenosis. Eur Heart J 1992; 13(Suppl): 82(Abstract).

]136] Gellman J, Ezekowitz MD, Sarembock IJ, et al. Effect of lovastatin on intimal hyperplasia after balloon angioplssty: a study in an atherosclerotic hypercholesterolemic rabbit. J Am Coll Cardiol 1991;17:251-259.

11371 Weintmub WS, Bocuzzi SJ, Klein JL, et al. Lack of effect of lovastatin on restenosis after coronary angioplasty. N Engl J Med 1994;331:1331-1337.

[I381 Leaf A, Weber PC. Cardiovascular effects of n-3 fatty acids. N Engl J Med 1990;318:549-557.

11391 Dehmer GJ, Popma JJ, Van Den Berg EK, et al. Reduction in the rate of early restenosis after coronary angioplasty by a diet supple- mented with n-3 fatty acids. N Engl J Med 1988;319:733-740.

]140] Mihter MR. Gallmo RA, Leffingwell A, et al. Usefulness of fish oil supplement in preventing clinical evidence of restenosis after per- cutaneous transluminal coronary angioplasty. Am J Cardiol 1989;64:294-299.

11411 Grigg LE. Kay TWH, Valentine PA, et al. Determinants of resteno- sis and lack of effect of dietary supplementation with eicosapenta- enoic acid on the incidence of coronary artery restenosis after angioplasty. J Am Coll Cardiol 198913665-672.

[I421 Reis GJ, Boucher TM, Sipperly ME, et al. Random&d trial of fish oil for prevention of restenosis after coronary angioplasty. Lancet 1989;2:177-181.

[143] Liu MW, Roubin GS, Robinson KA, et al. Trapidil in preventing restenosis after balloon angioplasty in the atherosclerotic rabbit. Circulation 1990;81:1089-1093.

[I441 Okamoto S, Masaaki I, Setsuda M, Konishi T, Nakano T. Effects of trapidil (triazolopyrimiclme), a platelet-derived growth factor (PDGF) antagonist in preventing restenosis after percutaneous transluminal coronary angioplasty. Am Heart J 1992;123: 1439- 1444(AbstractJ.

[I451 STARC study group. Trapidil (platelet-derived growth factor in- hibitor) prevents restenosis after PTCA: results of the STARC study. Eur Heart J 1993; I Nabstr Suppl):277.

Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739by gueston 18 February 2018

Page 12: Mechanisms and prevention of restenosis: from experimental

846 C. Bauters et al./ Cardiovascular Research 31 (19%) 835-846

[ 1461 Lundergan C, Foegh ML, Vargas R, et al. Inhibition of myointimal proliferation of the rat carotid artery by the peptides, angiopeptin and BIM 23034. Arteriosclerosis 1989;80:49-55.

[147] Santoian EC, Schneider JE, Gravanis MB, et al. Angiopeptin inhibits intimal hyperplasia after angioplasty in porcine coronary arteries. Circulation 1993;88: 1 1 - 14.

[ 1481 Hierowski MT, Liebow C. DuSpain K, Schally AV. Stimulation by somatostatin of dephosphorylation of membrane proteins in pancre- atic cancer MIA PaCa-2 cell line. FEBS Lett 1985;179:252-256.

[I491 Bauters C, Van Belle E, Wemert N, et al. Angiopeptin inhibits oncogene induction in rabbit aorta after balloon denudation. Circu- lation 1994892327-2331.

[150] Emanuellson H, Beatt KJ, Bagger JP, et al. Long-term effects of angiopeptin treatment in coronary angioplasty. Reduction of clini- cal events but not angiographic restenosis. Circulation 1995;91:1689-1696.

[ 1511 Kent KM, Williams DO, Cassagneau B, et al. Double-blind, con- trolled trial of the effect of angiopeptin on coronary restenosis following balloon angioplasty. Circulation 1993;88(Suppl I>:I- 506(Abstract).

[152] Whitworth HB, Roubin OS, Hoilman J, et al. Effect of nifedipine on recurrent stenosis after percutaneous transluminal coronary an- gioplasty. J Am Co11 Cardiol 1986;8:1271-1276.

[153] Corcos T, David PR, Val PG, et al. Failure of dihiazem to prevent restenosis after percutaneous transluminal coronary angioplasty. Am Heart J 1985;109:926-931.

[ 1541 Pepine CJ, Hirshfeld JW, McDonald RG, et al. A controlled trial of corticosteroids to prevent restenosis after coronary angioplasty. Circulation 1!3!%;81:1753-1761.

[155] Grines CL, Rizik D, Levine A, et al. Colchicine angioplasty restenosis trial (CART). Circulation 1991;84:II-365cAbstract).

[ 1561 ACCORD Study Group. Nitric oxide donors reduce restenosis after coronary angioplasty. J Am Coil Cardiol 1994,23(Suppl):59A(Ab- stract).

[I571 Schwartz RS, Murphy JG, Edwards WD, Cammd AR, Vlietstra RE, Holmes DR. Restenosis after balloon angioplasty: a practical proliferative model in porcine coronary arteries. Circulation 1990;82:2190-2200.

[I581 Karas SP, Gravanis MB, Robinson KA, Anderberg A, Heam JA, King SB. Comparison of the response to coronary artery balloon injury and stenting in swine: an animal model of restenosis. Circulation 1990,82(Suppl III):III-497(Abstract).

[159] Van Belle E, Bauters C, Wemert N, et al. Angiotensin converting enzyme inhibition prevents proto-oncogene expression in the vas- cular wall after injury. J Hypertension 1995; 13: 105 112.

[160] Serruys PW, de Jaegere P, Kiemeneij F, et al. A comparison of baBoon-expandable-stent implantation with balloon angioplasty in patients with coronary artery disease. N Engl J Med 1994;331:489- 495.

[161] Fischman DL, Leon MB, Bairn DS, et al. A randomized compari- son of coronary-stent placement and balloon angioplasty in the treatment of coronary artery disease. N Engl J Med 1994;331:496- 501.

[162] Wolinski H, Thung SN. Use of a perforated balloon catheter to deliver concentrated heparin into the wall of the normal canine artery. J Am Co11 Cardiol 1!%0;15:475-481.

(1631 Fram DB, Aretz TA, Mitchel JF, et al. Localized intramural delivery of a marker agent during balloon angioplasty: a new technique using hydrogel-coated balloons and active diffusion. Circulation 1992;86+38dAbstractJ.

[164] Camenzind E, Reys A, Ligduut J, et al. Local intracoronary

delivery of heparht and of a somatostatin analogue (octreotide) following angioplasty in man: evaluation of efficacy of delivery by radio-isotopic technique. J Am Co11 Cardiol 1995;Abstract Supple- meot:285A(Abstract).

[I651 Hong MK, Bhatti T, Matthews BJ, et al. Locally delivered an- giopeptin reduces intimal hypetplasia following balloon injury in rabbits. Circulation 1991;84:11-7xAbstract).

[166] Wilenski RL, Match KL, Hathaway DR. Direct intraarterial wall injection of microparticles via a catheter: a potential drug delivery strategy following angioplasty. Am Heart J 1991;122:1136-1141.

[I671 March KL, Mohanraj S, Ho P, Wilenski RL, Hathaway DR. Biodegradable microspheres containing a colchicine analog inhibit DNA synthesis in vascular smooth muscle cells. Circulation 19!n;86:1-381(Abstract).

[168] Steg PG, Feldman L, Scoazec JY, et al. Percutaneous delivery of adenovims for arterial gene transfer of endothelial and smooth muscle cells. Circulation 199490: 1648-1656.

[I691 Von der Leyen H. Gibbons GH, Morishita R, et al. In vivo gene transfert of nitric oxide synthase inhibits neointima formation in injured rat carotid arteries. Eur Heart J 1994,15:426(Abstract).

[170] Asahara T, Bauters C, Pastore C, et al. Local delivery of vascular endothelial growth factor accelerates reendothelialization and atten- uates intimal hyperplasia in balloon-injured rat carotid artery. Cir- culation 1995;91:2793-2801.

[171] Bertrand ME, Lablanche JM, Leroy F. et al. Percutaneous transhr- minal coronary rotary ablation with rotablator (European experi- ence). Am J Cardiol 1992.69470-478.

[172] Buchbinder M, Leon M, Wartb D, et al. Multi-center registry of percutaneous coronary rotational ablation using the rotablator. J Am Coil Cardiol 1992; 19:33A(Abstmct).

[I731 Eigler N, Forrester JS. Nonpharmacologic device prevention of coronary restenosis. In: Topol, EJ, ed. Textbook of Iinterventional Cardiology. Philadelphia: WB Saunders, 1994400-414.

[174] Safian RD. Gelbfish JS, Emy RE, et al. Coronary atherectomy: clinical, angiographic, and histological findings and observations regarding potential mechanisms. Circulation 1990,82:69-75.

[ 1751 Top01 EJ, Leya F, Pinkerton CA, et al. A comparison of directional atherectomy with coronary angioplasty in patients with coronary artery disease: the CAVEAT Study Group. N Engl J Med 1993;329:221-227.

[176] Elliott JM, Berdan LG, Holmes DR, et al. One-year follow-up in the coronary angioplasty versus excisional atherectomy trial (CAVEAT I). Circulation 1995;91:2158-2166.

[I771 S&wart U, Puel J, Mirkovitch V, Joffre F, Kappenberger L. humvascular stems to prevent occlusion and restenosis after trans- huninal angioplasty. N Engl J Med 1987;316:701-706.

[178] Serruys PW, Strauss BH, Beatt KJ, et al. Quantitative follow-up after placement of a self-expanding coronary stent. N Engl J Med 1991;324:13-17.

[179] Mintz GS, Popma JJ, Pichard AD, et al. Differing mechanisms of late arterial responses to transcatheter therapy: a serial quantitative angiographic and intravascular ultrasound study. Eur Heart J 1995;Abstract Supplement:385(Abstract).

[ 1801 Dc Scheerder I, Wilczek K, Van Dorpe J, et al. Angiopeptin loaded stcnts inhibit the neointimal reaction induced by polymer coated stems implanted in porcine coronary arteries. Circulation 1994,90:1-597cAbstract).

[I811 Bauters C, Lablanche JM, Renaud N, McFadden EP, Hamon M, Bertrand ME. Morphologic changes after percutaneous translumi- nal coronary angioplasty of unstable plaques. Insights from serial angioscopic follow-up. Eur Heart J in press.

Downloaded from https://academic.oup.com/cardiovascres/article-abstract/31/6/835/375739by gueston 18 February 2018