stent-based immunosuppressive therapies for the prevention of restenosis

10
Stent-based immunosuppressive therapies for the prevention of restenosis Meenakshi Aggarwal a , Philip S. Tsao a , Alan Yeung a , Andrew J. Carter b, * a Stanford University Medical Center, Stanford, CA, USA b Interventional Cardiology Research, Providence Heart Institute, Providence St. Vincent Medical Center, Providence Health System, 9205 Southwest Barnes Road, Portland, OR, 97225, USA Received 11 June 2003; received in revised form 31 July 2003; accepted 31 July 2003 1. Introduction The long-term clinical efficacy of intracoronary stenting is limited by restenosis, which occurs in 15% to 30% of patients [1,2]. In-stent restenosis is due solely to neointimal hyperplasia [3 – 6]. Stent-induced mechanical arterial injury and a foreign body response to the prosthesis incites acute and chronic inflammation in the vessel wall (Fig. 1). The subsequent elaboration of cytokines and growth factors induces multiple signaling pathways via mTOR to activate smooth muscle cell migration and proliferation. The expres- sion of inflammatory cytokines from proliferating T cells and macrophages are associated with the release of growth factors, such as platelet-derived growth factor, basic fibro- blast growth factor, endothelial cell growth factor, or trans- forming growth factor-beta, which induce smooth muscle cell proliferation and migration as well as secretion of matrix proteins. The long-term effects of smooth muscle cell migration, proliferation and matrix formation is the development of neointimal hyperplasia that may obstruct the stent lumen resulting in restenosis. Thus, pharmacolog- ical compounds or other agents that target inflammation and cellular proliferation may be ideal candidates as stent-based therapies for the prevention of restenosis. The promising early clinical results of the potent stent- based immunosuppressive therapy, sirolimus, lead a number of investigators to explore the effects of other immunosup- pressive agents as stent-based therapies for the prevention of restenosis. The purpose of this manuscript is to review the relevant preclinical and clinical data in the field of stent- based immunosuppressive therapies for the prevention of restenosis. An understanding of the physical properties and pharmacological effects of these compounds as well as the proposed drug delivery stent systems will improve our interpretation of clinical outcomes with this class of drug- eluting stents. The reader will appreciate that a ‘‘class effect’’ is unlikely to be observed as these immunosuppres- sive compounds each with uniquely different molecular weight, solubility, and mechanism of action (Table 1) are explored as stent based therapies for the prevention of restenosis. This information will also serve as a helpful comparison to other potentially effective stent-based thera- pies such as paclitaxel. 2. Dexamethasone Dexamethasone is a potent glucocorticoid with anti- inflammatory and antiproliferative properties. In vitro stud- ies with cultured bovine [7] and human [8] smooth muscle cells demonstrated that steroids are effective in suppressing smooth muscle cell proliferation. The mechanism of action by which steroids inhibit smooth muscle cell proliferation is multifold. Steroids by virtue of their anti-inflammatory property reduce collection of inflammatory cells at the site of vessel wall trauma. The steroid induced anti-inflamma- tory actions include inhibition of leukocyte adhesion to endothelial cells, reduction in leukocyte aggregation, sup- pression of platelet derived growth factor, and reduced production of cytokines, nuclear proteins, fibroblasts and macrophages [9]. In vivo experiments with systemic admin- istration of glucocorticoids have demonstrated reduction in intimal hyperplasia [10] and atherosclerotic plaque forma- tion [11]. Unfortunately, these results have not been repro- duced in human clinical trials [12,13], most likely due to inadequate concentration of drug at the intervention site, thus, suggesting the need for more effective local drug delivery. The human clinical trials failed to demonstrate a reduction in post-PTCA restenosis by systemic administra- tion of steroids for 1 or 7 days. The prolonged and high dose 1522-1865/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved. doi:10.1016/S1522-1865(03)00165-3 * Corresponding author. Interventional Cardiology Research, Provi- dence Heart Institute, Providence St. Vincent Medical Center, Providence Health System, 9205 Southwest Barnes Road, Portland, OR, 97225, USA. Tel.: +1-503-216-5206. E-mail address: [email protected] (A.J. Carter). Cardiovascular Radiation Medicine 4 (2003) 98 – 107

Upload: meenakshi-aggarwal

Post on 20-Sep-2016

214 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Stent-based immunosuppressive therapies for the prevention of restenosis

Cardiovascular Radiation Medicine 4 (2003) 98–107

Stent-based immunosuppressive therapies for the prevention

of restenosis

Meenakshi Aggarwala, Philip S. Tsaoa, Alan Yeunga, Andrew J. Carterb,*

aStanford University Medical Center, Stanford, CA, USAb Interventional Cardiology Research, Providence Heart Institute, Providence St. Vincent Medical Center,

Providence Health System, 9205 Southwest Barnes Road, Portland, OR, 97225, USA

Received 11 June 2003; received in revised form 31 July 2003; accepted 31 July 2003

1. Introduction

The long-term clinical efficacy of intracoronary stenting

is limited by restenosis, which occurs in 15% to 30% of

patients [1,2]. In-stent restenosis is due solely to neointimal

hyperplasia [3–6]. Stent-induced mechanical arterial injury

and a foreign body response to the prosthesis incites acute

and chronic inflammation in the vessel wall (Fig. 1). The

subsequent elaboration of cytokines and growth factors

induces multiple signaling pathways via mTOR to activate

smooth muscle cell migration and proliferation. The expres-

sion of inflammatory cytokines from proliferating T cells

and macrophages are associated with the release of growth

factors, such as platelet-derived growth factor, basic fibro-

blast growth factor, endothelial cell growth factor, or trans-

forming growth factor-beta, which induce smooth muscle

cell proliferation and migration as well as secretion of

matrix proteins. The long-term effects of smooth muscle

cell migration, proliferation and matrix formation is the

development of neointimal hyperplasia that may obstruct

the stent lumen resulting in restenosis. Thus, pharmacolog-

ical compounds or other agents that target inflammation and

cellular proliferation may be ideal candidates as stent-based

therapies for the prevention of restenosis.

The promising early clinical results of the potent stent-

based immunosuppressive therapy, sirolimus, lead a number

of investigators to explore the effects of other immunosup-

pressive agents as stent-based therapies for the prevention of

restenosis. The purpose of this manuscript is to review the

relevant preclinical and clinical data in the field of stent-

based immunosuppressive therapies for the prevention of

restenosis. An understanding of the physical properties and

1522-1865/03/$ – see front matter D 2003 Elsevier Inc. All rights reserved.

doi:10.1016/S1522-1865(03)00165-3

* Corresponding author. Interventional Cardiology Research, Provi-

dence Heart Institute, Providence St. Vincent Medical Center, Providence

Health System, 9205 Southwest Barnes Road, Portland, OR, 97225, USA.

Tel.: +1-503-216-5206.

E-mail address: [email protected] (A.J. Carter).

pharmacological effects of these compounds as well as the

proposed drug delivery stent systems will improve our

interpretation of clinical outcomes with this class of drug-

eluting stents. The reader will appreciate that a ‘‘class

effect’’ is unlikely to be observed as these immunosuppres-

sive compounds each with uniquely different molecular

weight, solubility, and mechanism of action (Table 1) are

explored as stent based therapies for the prevention of

restenosis. This information will also serve as a helpful

comparison to other potentially effective stent-based thera-

pies such as paclitaxel.

2. Dexamethasone

Dexamethasone is a potent glucocorticoid with anti-

inflammatory and antiproliferative properties. In vitro stud-

ies with cultured bovine [7] and human [8] smooth muscle

cells demonstrated that steroids are effective in suppressing

smooth muscle cell proliferation. The mechanism of action

by which steroids inhibit smooth muscle cell proliferation is

multifold. Steroids by virtue of their anti-inflammatory

property reduce collection of inflammatory cells at the site

of vessel wall trauma. The steroid induced anti-inflamma-

tory actions include inhibition of leukocyte adhesion to

endothelial cells, reduction in leukocyte aggregation, sup-

pression of platelet derived growth factor, and reduced

production of cytokines, nuclear proteins, fibroblasts and

macrophages [9]. In vivo experiments with systemic admin-

istration of glucocorticoids have demonstrated reduction in

intimal hyperplasia [10] and atherosclerotic plaque forma-

tion [11]. Unfortunately, these results have not been repro-

duced in human clinical trials [12,13], most likely due to

inadequate concentration of drug at the intervention site,

thus, suggesting the need for more effective local drug

delivery. The human clinical trials failed to demonstrate a

reduction in post-PTCA restenosis by systemic administra-

tion of steroids for 1 or 7 days. The prolonged and high dose

Page 2: Stent-based immunosuppressive therapies for the prevention of restenosis

Fig. 1. Photomicrographs of a human coronary artery after stent placement.

Note the severe inflammatory and foreign-body response to the stent with

diffuse inflammatory cell infiltration, and multiple giant cells (courtesy of

Dr. Andrew Farb, Armed Forces Institute of Pathology, Washington, DC).

Table 1

General mechanisms of action of immunosuppressive drugs

Small Molecules

CsA, tacrolimus (FK506)

Inhibition of calcineurin phosphatase

Mycophenolate mofetil (MMF)

Inhibition of inosine monophosphate dehydrogenase (IMPDH)

Sirolimus

Inhibition of mTOR1 and 2

Steroids

Pleiotropic effects including blocking activation of nuclear factor-

kappa B (NF-nB)

CsA, cyclosporine.

M. Aggarwal et al. / Cardiovascular Radiation Medicine 4 (2003) 98–107 99

systemic intravenous and intramuscular administration of

glucocorticoids in humans is limited by potential for side

effects. Consequently, dexamethasone-eluting stents are a

prospective technique to allow for local delivery of high

concentration of glucocorticoids while limiting their sys-

temic side effects.

Lincoff et al. [14] are one of the first groups to study the

efficacy of intracoronary delivery of dexamethasone via a

polymer-coated dexamethasone-eluting stent in a porcine

coronary injury model. A 125-Am diameter tantalum wire

configured into a 16 mm long balloon expandable coil stent

was used. Each stent was sprayed and coated with a mixture

of poly-L-lactic acid (PLLA) and dexamethasone in a 2:1

ratio with 0.8 mg of dexamethasone and 0.4 mg of PLLA

per stent. Two different formulations of PLLAwere used—a

low molecular weight polymer of 80 kD and a high

molecular weight polymer of 321 kD. At 28 days after

implant, the stents coated with low molecular weight PLLA

produced a severe inflammatory response at the polymer

tissue interface. This infiltrate consisted of mononuclear

cells, lymphocytes and multinucleated giant cells. On the

contrary, arteries with high molecular weight PLLA stents

did not demonstrate an acute or chronic inflammatory

response. Nonetheless, neither high molecular weight

PLLA nor combination of high molecular weight PLLA–

dexamethasone resulted in a decrease in neointimal hyper-

plasia. The proportion of dexamethasone initially contained

within the stent coating was estimated in the arterial wall with

8% at 1 h and 10% at 24 h after implantation. Dexamethasone

concentration in the arterial tissue was 90,000 to 300,000

times higher than the serum levels at 24 h after stent

implantation. At 28 days, the arterial tissue dexamethasone

concentration remained 3000 times higher relative to the

serum levels. This study demonstrated that dexamethasone

could be delivered via a polymer-coated stent in a safe and

sustained manner though the dose appeared insufficient to

produce a reduction in neointimal hyperplasia [14].

Strecker et al. [15] evaluated the effect of polymer-coated

dexamethasone eluting stents on neointimal hyperplasia in a

canine model. Balloon expandable flexible tantalum stents

were coated with a pure polylactide (dl-PLA) or a polylac-

tide-co-polymer (PLA-Co-TMC). The ratio of polymer to

dexamethasone was 84:16 with each stent containing ap-

proximately 4 mg of dexamethasone per cm stent coating.

Digital subtraction angiography (DSA) was performed at 3,

6, 9, 12 and 24 weeks after stent implantation. Arteries with

dexamethasone coated stents demonstrated about 30% less

neointimal thickness within the stented area at 24 weeks

when compared with noncoated stents (183 vs. 263 Am). On

angiography, dexamethasone-coated stents revealed 29%

less stenosis than noncoated stents at 24 weeks.

More recent data has demonstrated that methylpredniso-

lone-coated stents decrease neointimal hyperplasia in a

porcine coronary model [16]. Ten percent (g/g) of methyl-

prednisolone in a polyfluoroalkoxy phosphazene polymer

(PFM-P75) was sprayed onto the surface of stainless steel

balloon expandable coronary stents. A barrier coat of

1% (g/v) PFM-P75 was then applied to allow sustained

release of methylprednisolone. At 4 weeks, the coronary

arteries with methylprednisolone eluting stents had a de-

creased inflammatory score surrounding the stent filaments

(0.46 + 0.54 vs. 2.34 + 0.75, P < .001) and significantly less

neointimal hyperplasia vs. controls (2.53 + 0.85 vs.

4.29 + 1.28 mm2, respectively). In vitro methylpredniso-

lone-release curves revealed 50% methylprednisolone re-

lease from the stent within 33 days. In summary, the studies

by Lincoff et al. [14], Strecker et al. [15] and Ping et al.

[16] provide preliminary data that steroid-eluting stents are

safe and may inhibit in-stent neointimal hyperplasia. This

preclinical work, while inconclusive, nonetheless has set

the stage for clinical trials of steroid eluting stents.

Study of Antirestenosis with the Biodivysio Dexameth-

asone Eluting Stent, STRIDE, is a multicenter trial evalu-

ating the safety and efficacy of Biodivysio Drug Delivery

Phosphorylcholine Coated (DD PC) stent with dexa-

methasone in de novo coronary lesions [17]. Seventy-one

patients at a mean age of 61.9 years were enrolled at eight

centers in Belgium. Their clinical profile consisted of 63%

having hyperlipidemia, 56% had hypertension, 41% had

history of previous MI and 28% had unstable angina

pectoris. The Biodivysio DD PC stent was dipped in a

15 mg/ml dexamethasone solution for at least 5 min and air-

dried for 5 min, resulting in 45 Ag of dexamethasone per

mm stent. Three different lengths of stents were available—

11, 15 or 18 mm. Patients received ASA indefinitely and

Page 3: Stent-based immunosuppressive therapies for the prevention of restenosis

M. Aggarwal et al. / Cardiovascular Radiation Medicine 4 (2003) 98–107100

ticlodipine 250 mg/day for 28 days. The primary endpoint

was angiographic restenosis at 6 months with secondary

endpoints of major adverse cardiac events at 30 days and

6 months. Forty-one percent of the treated lesions were in

the LAD and 30% in the RCA. Preliminary results have

revealed four major adverse cardiac events at 30 days (one

in hospital death secondary to stent thrombosis, one non-

stent cardiac death, one non-Q wave MI, and one recurrence

of chest pain requiring target vessel revascularization).

Long-term clinical follow-up is in progress, although pre-

liminary data indicates only a modest reduction of angio-

graphic late lumen loss for patients with dexamethasone-

eluting stents at 6 months. Interestingly, the authors

reported potentially more favorable effects for dexametha-

sone-eluting stents in patients with unstable angina as

compared to those with stable angina at the time of initial

revascularization procedure. Future prospective, random-

ized studies with dexamethasone eluting stents are planned

to confirm these initial findings (BRILLIANT).

3. Sirolimus

Sirolimus (formerly rapamycin, Wyeth Ayerst) is a natu-

rally occurring macrolide antibiotic. It was originally

extracted from soil in Easter Island. It is produced by natural

fermentation from the fungus Streptomyces. Sirolimus is a

potent immunosuppressive agent, approved by the FDA in

1999 for treating allograft rejection in renal transplant

patients. More recently, the antiproliferative effects of siroli-

mus were recognized leading to its use in drug-coated stents.

In theory, sirolimus has a dual mechanism of action to

potentially inhibit neointimal formation and reduce resteno-

sis (Fig. 2). Sirolimus has both potent anti-inflammatory and

antiproliferative properties. Initial studies have demonstrated

Fig. 2. This figure illustrates the fundamental mechanism of action for t

that sirolimus is a cytostatic inhibitor of cytokine and growth

factor mediated cell proliferation [18,19]. Sirolimus inhibits

the cell cycle at the G1/S phase by binding to a cellular

receptor FKBP12 which in turn blocks molecular target of

rapamycin (mTOR) activation. Consequently, down regula-

tion of the cyclin dependent kinase p27kip1 is prevented and

phosphorylation of the retinoblastoma protein is inhibited.

Experimental data by Marx et al. [20], Poon et al. [21] and

Gallo et al. [22] has helped to demonstrate the mechanism of

action of sirolimus on vascular smooth muscle cells. Marx

et al. [20] demonstrated sirolimus mediated inhibition of rat

and human vascular smooth muscle cell proliferation in

vitro. Gallo et al. [22] demonstrated that systemic adminis-

tration of sirolimus was effective in reducing neointimal

hyperplasia in a porcine balloon injury model. In addition to

the biological properties of sirolimus, its physical properties

are ideal for the application as a locally delivered agent.

Sirolimus is a hydrophobic compound with a molecular

weight of 942 and thus, low solubility in aqueous solutions.

Due to its lipophilicity, the drug passes easily through the

cell membrane allowing for intramural distribution and

prolonged arterial tissue retention. Together the structural

properties and biological effects of sirolimus suggest that the

compound may be ideally suited for stent-based delivery in

the prevention of restenosis.

Experimental studies were implemented in 1998 to

determine the feasibility and efficacy of sirolimus-eluting

stents. In 1999, we performed preclinical studies to deter-

mine the efficacy of sirolimus-eluting stents alone or in

combination with dexamethasone to reduce in-stent neo-

intimal hyperplasia using a porcine coronary model [23]. At

28 days, a 50% reduction in neointimal proliferation was

demonstrated with sirolimus-eluting stents compared to

bare metal, P < .0001. The combination of sirolimus with

dexamethasone failed to produce a synergistic effect. The

he anti-proliferative and immunosuppressive compound sirolimus.

Page 4: Stent-based immunosuppressive therapies for the prevention of restenosis

M. Aggarwal et al. / Cardiovascular Radiation Medicine 4 (2003) 98–107 101

reduction in neointimal hyperplasia translated to a signifi-

cant reduction in in-stent restenosis for sirolimus-eluting

stents compared to bare metal (26F 11% vs. 55F 20%,

respectively). Sirolimus alone or in combination with dexa-

methasone profoundly suppressed strut-associated inflam-

mation. Endothelialization scores were the same for both

the sirolimus and metal stents. Arterial wall protein expres-

sion at 7 days, assessed by Western blot, revealed a

profound reduction in PCNA expression and pRb phos-

phorylation with sirolimus-eluting stents. These molecular

effects of sirolimus result in inhibition of neointimal hyper-

plasia. In addition, we demonstrated a 70% reduction in the

inflammatory cytokine MCP-1 with sirolimus-eluting stents

(Fig. 3). This preclinical study showed that sirolimus-

eluting stents could significantly reduce in-stent restenosis

by cytostatic inhibition of the cell cycle and reduction in

inflammatory cytokines.

The clinical trials with sirolimus-eluting stents have

confirmed its success in reducing in-stent restenosis. The

First-In-Man trial with sirolimus-eluting stents was con-

ducted by Sousa et al. [24,25] to assess the safety and

feasibility of the sirolimus eluting Bx Velocity stent in the

treatment of de novo coronary lesions. Forty-five patients

with stable angina were enrolled at two centers—Sao Paulo,

Brazil (30 patients) and Rotterdam, Netherlands (15 patients).

Patients were treated with either a 3.0 or 3.5 mm diameter,

18 mm long, fast release (FR) or slow release (SR) sirolimus-

eluting stent (15 FR and 15 SR in Brazil and 15 SR in

Rotterdam). The FR formulation delivered the drug in less

than 15 days and the SR formulation delivered the drug in

90 days. All patients received ASA indefinitely and clopi-

dogrel for 60 days. Coronary angiography and IVUS were

performed immediately after the procedure, at 4, 12 and at

24 months. The angiographic and IVUS results at 4 and

12 months showed 0% binary restenosis. One year in-stent

MLD (FR, 2.73F 0.3 mm and SR, 2.87F 0.4 mm) and

Fig. 3. Western blot of porcine coronary artery specimens at 7 days after

placement of bare metal (St), sirolimus (SRL) and dexamethasone (Dex)

eluting stents. MCP-1 expression is not detectable in noninjured aorta (A).

The relative expression of MCP-1 is significantly less for sirolimus treated

arteries in comparison with arteries treated with bare metal stents.

%DS (FR, 8.9F 6.1% and SR, 6.7F 7%) remained essen-

tially unchanged compared to 4-month follow-up. Sousa

et al. [24,25] recently reported the 24-month clinical, angio-

graphic and IVUS follow-up for the 30 patients who received

either the FR release or SR formulation in Sao Paulo, Brazil.

After 2 years, no patient developed in-stent restenosis, and

90% were free of repeat target vessel revascularization. No

patient deaths occurred during the study period. One patient

had myocardial infarction at 14 months secondary to target

vessel occlusion, which prevented further angiographic as-

sessment. Angiographic assessment was deferred in another

patient due to developing pneumonia the week prior to

scheduled angiography. Angiography and IVUS in 28 of

30 patients demonstrated a similar MLD, % diameter steno-

sis and neointimal hyperplasia volume at 24 months in

comparison with 12 months. In-lesion and in-stent MLD

were greater in the SR group than the FR group while plaque

volume was similar for each group by IVUS. In the SR

group, in-stent and in-lesion MLD increased at 2 years vs.

1 year. No patient had in-stent restenosis (z50% diameter

stenosis). This is the longest follow up to date available on

sirolimus-eluting stents in man. These preliminary results did

not show a ‘‘late catch up’’ in restenosis and in fact suggest a

sustained effect of stent-based sirolimus in reducing reste-

nosis. These promising first-in-man results encouraged con-

firmation with randomized, placebo-controlled, multicenter

clinical trials.

The Randomized study with the sirolimus-coated Bx

VElocity balloon expandable stent in the treatment of

patients with de novo native coronary Lesions (RAVEL)

was a prospective, multicenter, randomized, double blind

clinical trial comparing bare metal and sirolimus-coated

stents [26]. This trial enrolled 238 patients from 15 centers

in Europe and 4 centers in Latin America. The patients were

randomized to a single sirolimus-coated stent (140 Ag/cm2)

or the bare metal Bx Velocity stent. The primary endpoint

was minimum lumen diameter measured by quantitative

coronary angiography (QCA) at 6-months. Prerequisite

native vessel diameter was 2.5 to 3.5 mm. Baseline demo-

graphics, anginal status and pre- and postprocedural vessel

diameter in both the sirolimus and control group were

similar. QCA analysis within the stent at 6 months demon-

strated 0% restenosis rate in the sirolimus-treated group

compared to 26% in the control, P < .0001. Late loss in the

sirolimus-treated group was a mere 0.01 mm compared to

0.80 mm for control, P < .0001. This resulted in a MLD at

follow up of 2.42 mm for the sirolimus treated group vs.

1.64 mm for control. No major adverse cardiac events were

seen in 96.7% of the sirolimus-treated group whereas only

72.9% of the bare metal group was event free. Serruys et al.

recently demonstrated that sirolimus-eluting stents inhi-

bited restenosis irrespective of the vessel size. In a subset

of 95 patients (48 sirolimus-eluting stents, 47 bare metal

stents), a motorized intravascular ultrasound pullback

along with quantitative coronary analysis was performed

at 6 months. There was no evidence of edge effect as

Page 5: Stent-based immunosuppressive therapies for the prevention of restenosis

M. Aggarwal et al. / Cardiovascular Radiation Medicine 4 (2003) 98–107102

documented by intravascular ultrasound analysis in 95 of

the 237 patients or as demonstrated by QCA at 6 months.

Data from the subset of patients undergoing intravascular

ultrasound was reviewed to assess the incidence of incom-

plete stent apposition (ISA) at 6 months. ISA was defined

as z1 strut clearly separated from vessel wall with evi-

dence of blood speckles behind the strut. The incidence of

ISA in patients receiving sirolimus-eluting stent was 20% at

6 months compared to 4% in the group receiving a bare metal

stent (P < .015) with diabetics being more prone to ISA.

Nonetheless, ISA was not associated with adverse clinical

events. It is unclear whether ISA is the result of late acquired

malapposition or a consequence of an acute incomplete

deployment. In this trial, IVUS was not performed at time

of stent deployment, thereby making it difficult to determine

the underlying mechanism. After 1 year, a 94.2% event free

survival (freedom from death, myocardial infarction, target

vessel revascularization, target lesion revascularization) was

reported for the sirolimus-treated group compared to 71.2%

for the control group. These results set a stage for larger

randomized trials to be conducted in the United States.

SIROLIMUS-Coated BX Velocity Balloon-Expandable

Stent in the Treatment of Patients with De Novo Coronary

Artery Lesions (SIRIUS study) is a prospective, multicenter,

randomized, double-blind clinical trial that was conducted

in 55 centers in the United States. Eleven hundred and one

patients with focal de novo native coronary arterial lesions

(2.5–3.5 mm diameter, 15 to 30 mm long) were randomized

to treatment with sirolimus-coated (109 Ag/cm2) or baremetal

Bx Velocity stents between February and August 2001.

Patients received ASA indefinitely and either clopidogrel or

ticlodipine for 3 months postprocedure. The primary end-

point of the SIRIUS study was target vessel failure (cardiac

death, myocardial infarction, target lesion revascularization)

at 9 months postprocedure. Secondary endpoints included

angiographic in-stent and in-segment binary restenosis at

8 months, angiographic in-stent and in-segment minimum

lumen diameter at 8 months, major adverse cardiac events

up to 5 years and economic factors (index hospitalization

costs, length of stay and repeat hospitalizations) for up to

12 months. This trial differs from RAVEL in that it was a

larger cohort of patients, involved longer lesions, allowed

two stents per lesion, required a longer duration of anti-

platelet therapy (3 months vs. 2 months for RAVEL) and

will provide economic data. The results of the SIRIUS trial

were presented during the XIIIth Annual Transcatheter

Cardiovascular Therapeutics, September 2002 [27].

Patient demographics were similar in both groups with

respect to age, hypertension, hyperlipidemia and diabetes.

Approximately 60% of the patients had single vessel dis-

ease, 40% a lesion in the left anterior descending coronary

artery, and 60% of the lesions were classified as ACC/AHA

lesion class B2 or C. The average lesion length was 14.3 mm

for the sirolimus-treated group and 14.6 mm for the bare

metal group. On average, 1.4 stents were implanted per

patient with overlapping of stents in approximately 25% of

cases. Quantitative angiographic analysis of the in-stent

segment at 8 months demonstrated a late loss of 0.14 mm

for the sirolimus-eluting group compared to 0.92 mm for the

bare metal group, resulting in an 85% reduction in late loss

and 94% decline in in-stent restenosis, P < .001. Quantita-

tive angiographic analysis of the treated segment (stent plus

5 mm proximal or distal) demonstrated a 67% reduction in

late loss and a 72% decrease in restenosis for the sirolimus-

eluting group in comparison with the BX Velocity, P < .001.

Thus, quantitative angiographic analysis of the treated

segment at 8 months demonstrated less effective suppres-

sion of neointimal hyperplasia at both the proximal and

distal peri-stent area compared to in-stent suppression of

neointimal hyperplasia by sirolimus-eluting stents. This

resulted in restenosis at the stent margins, which, in partic-

ular at the proximal margin, was not significantly different

between the sirolimus and control group. Further subgroup

analysis by reference vessel size revealed that smaller

vessels (2.3 mm diameter) were more prone to restenosis at

the proximal edge. There were two cases of stent thrombosis

in the sirolimus-treated group (0.4%) with four episodes of

stent thrombosis in the control group (0.8%). The primary

endpoint of target vessel failure (TVF) at 270 days was

8.6% for the sirolimus group compared to 21.0% for the

bare metal, P =.017, resulting in a reduction of TVF by 59%

for sirolimus treated patients. Clinical events of death and

MI were similar between the two groups at 270 days but

target lesion revascularization was decreased by 72% in the

sirolimus group, P < .001.

The larger sample size of the SIRIUS trial enabled

subgroup analysis of outcomes for specific lesion and patient

subsets. In general, the overall treatment effect of the

sirolimus-eluting stent was similar in several important

patient and anatomical subsets with a 75% relative reduction

in target lesion revascularization at 9-months. Within the

sirolimus group, target lesion revascularization was in-

creased in smaller vessels (2.3 mm or less) relative to

moderate (2.3 to 3.0 mm) or large vessels (3.0 mm or greater)

predominantly due to proximal margin peri-stent restenosis.

The frequency of target lesion revascularization was also

dependent on lesion length with an average frequency of

0.3 events per millimeter of stent length. In comparison with

BX Velocity, the sirolimus-eluting stent reduced the frequen-

cy of target lesion revascularization 70% independent of

lesion length. In diabetic patients the frequency of target

lesion revascularization was nearly twofold greater than

nondiabetic patients for sirolimus and bare metal stents.

Diabetic patients with reference vessel dimensions less than

2.3 mm had a 23% incidence of target lesion revasculariza-

tion in the sirolimus group as compared with 48% in the BX

Velocity group, P < .0001. From the various subgroup anal-

yses, it was concluded that sirolimus-eluting stents are safe

and effective in reducing neointimal hyperplasia in a more

complex group of patients and lesions. The suboptimal

efficacy in neointimal suppression at the proximal stent

margin in smaller vessels will require further evaluation and

Page 6: Stent-based immunosuppressive therapies for the prevention of restenosis

M. Aggarwal et al. / Cardiovascular Radiation Medicine 4 (2003) 98–107 103

perhaps modification of current stent deployment techni-

ques to include limiting the proximal zone of balloon injury

and insuring complete lesion coverage. Diabetic patients,

who benefited from sirolimus-eluting stents but to a lesser

extent than the nondiabetic subgroup, continue to pose a

challenge for durable long-term outcomes with stents. At

present, a dose escalation study, 3-D or Diabetic Double

Dose Study, is in-progress to determine if a 2X concentra-

tion of sirolimus (approximately 300 Ag/cm2 drug per stent

surface area) is safe and potentially more effective in the

challenging patient population.

With initial success in de novo native coronary lesions,

investigators have started to evaluate the role of sirolimus-

eluting stents in the management of in-stent restenosis

(ISR) [28,29]. Sousa et al. evaluated 30 patients, 16 in Brazil

and 14 in Netherlands, between February and May 2001 for

ISR in native coronary arteries. Patients were treated with

z1 sirolimus eluting Bx Velocity stent. Angiographic and

IVUS analysis was performed on day of implantation and at

6month follow up. Thirty patients received a total of 41 stents

with mean lesion length of 17.2F 5.7 mm. No acute post-

procedure cardiac events were noted. At 12 months, there

were no cases of stent thrombosis. Six-month data is under

evaluation. In contrast, Serruys et al. have suggested a

different response with a more frequent clinical event rate

including reintervention and stent thrombosis, particularly in

patients with more complex ISR as well as failed brachyther-

apy. Therefore, additional studies will be needed to determine

the utility of sirolimus-eluting stents for ISR and the relative

efficacy in comparison with vascular brachytherapy.

The utility of sirolimus-eluting stents in the treatment of

in-stent restenosis will be compared to endovascular bra-

chytherapy in a prospective multicenter randomized clinical

trial, the SISR study. The SISR study, A Multicenter,

Randomized Study of the Sirolimus-Eluting Bx Velocity

Balloon Expandable Stent vs. Intravascular Brachytherapy

in the Treatment of Patients with In-stent Restenosis, is

a 350-patient clinical trial comparing Cypherk sirolimus-

eluting stent placement to endovascular brachytherapy with

gamma or beta-emitting systems for ISR lesions in reference

vessels 2.75- to 3.75-mm diameter with lesion length of

<40 mm excluding chronic total occlusion. This pivotal

randomized clinical trial is designed to determine superior-

ity for Cypher sirolimus eluting stent by a reduction in 9-

month target vessel failure in comparison with endovascular

brachytherapy. The enrollment in the SISR trial is expected

to commence during the first quarter of 2003.

The Cypher sirolimus eluting is presently under investi-

gation in more complex lesion subsets typical of the ‘‘real

world’’ practice of interventional cardiology. At present,

registry studies are ongoing in patients with bifurcation

lesions, unprotected left main disease and acute myocardial

infarction. The efficacy of direct stenting with Cypher

sirolimus-eluting stents will be investigated in a randomized

clinical trial of 455 patients conducted in Canada and

Europe (E and C-SIRIUS). Multicenter randomized clinical

trials are planned for patients with multivessel coronary

disease amenable to stenting or CABG (ARTS II, FREE-

DOM) to determine equivalence of these revascularization

therapies. The FREEDOM study will be sponsored by the

NHLBI and conducted in 14 centers throughout the United

States to determine if multivessel Cypher sirolimus-eluting

stent placement provides similar outcomes to CABG at

3 years in diabetic patients with symptomatic coronary

artery disease. Ultimately, these studies will provide the

scientific evidence necessary to broaden clinical applica-

tions of sirolimus-eluting stents in the day-to-day practice

of interventional cardiology.

4. Tacrolimus

Tacrolimus is a potent immunosuppressant, which was

discovered in 1984 [30] and is approved by FDA for use in

management of liver transplantation and kidney allograft

rejection. Tacrolimus is a metabolite of the actinomycete

Streptomyces tsukubaensis. It is lipophilic and highly bound

to plasma proteins [32].

The cellular receptor, FKBP12, is the initial binding site

for both tacrolimus and sirolimus. However, unlike siroli-

mus, the tacrolimus–FKBP complex inhibits the activity of

calcineurin, a serine threonine phosphatase. The inhibition

of calcineurin leads to inactivation of transcription factors

(NF-AT) responsible for cytokine gene activation. Conse-

quently, inhibiting the transcription of many cytokines such

as interleukin (IL) 2, IL-3, IL-4, IL-5, tumor necrosis factor

alpha and granulocyte-macrophage colony-stimulating fac-

tor [31]. The potent inhibition of cytokine expression by

tacrolimus suggests a potential utility in the prevention of

restenosis. Mechanical injury to macrophage laden athero-

sclerotic plaques leads to cytokine gene expression by

macrophages and/or smooth muscles in the plaque [33].

The increase in cytokine level stimulates vascular smooth

muscle activation and proliferation ultimately leading to

neointimal formation and restenosis [33]. In addition, the

activated smooth muscle cells produce cytokines resulting

in a vicious cycle with continued stimulation of vascular

smooth muscle cell proliferation. The suppression of

cytokine production by tacrolimus may translate to a

reduction in in-stent restenosis. It is thought that tacrolimus

may have an advantage over other agents in that it may

preferentially inhibit the proliferation of vascular smooth

muscle cells without impairing endothelial cell regenera-

tion, thereby enabling stent integration into the arterial

wall. The local delivery of tacrolimus via a drug-eluting

stent should minimize the nephrotoxicity and neurotoxicity

associated with systemic administration of the agent while

allowing for the drug to be delivered to the intended site

of action.

Clinical trials with tacrolimus-eluting stents have been

initiated at the Heart Centre Sieburg, Germany. The PRES-

ENT study—PREliminary Safety Evaluation of Nanoporous

Page 7: Stent-based immunosuppressive therapies for the prevention of restenosis

M. Aggarwal et al. / Cardiovascular Radiation Medicine 4 (2003) 98–107104

Tacrolimus-eluting stents—is a two arm nonrandomized

prospective Phase I safety study comparing nanoporous

ceramic coated-stents with tacrolimus-eluting nanoporous

ceramic-coated stents. Tacrolimus at a dose of 60 Ag per

stent was used, to treat patients with a single focal native de

novo coronary arterial lesion. Preliminary data from this

study supports safety but suggests a need for additional dose

finding studies (E. Grube, MD, personal communication).

In addition to the aforementioned PRESENT trial,

JOMED is conducting the EVIDENT trial (Endo-Vascular

Investigation Determining the Safety of a New Tacrolimus

Eluting Stent Graft) to evaluate the safety of a tacrolimus-

eluting coronary stent graft for the treatment of saphenous

vein graft stenosis. The drug delivery platform is JOMED’s

balloon expandable PTFE coronary stent graft. The dose of

tacrolimus is 325 Ag per stent, which is significantly higher

than the 60 Ag dose in PRESENT trial. Professor Eberhard

Grube is also the principal investigator for the EVIDENT

trial, like PRESENT, which is being conducted at the Heart

Centre in Sieburg, Germany. To date, the preliminary results

of these trials indicates a lack of efficacy for this agent at the

present doses suggesting further dose finding or selection of

an alternative delivery method may be necessary to achieve

efficacy for this stent-based therapy.

5. Everolimus

Everolimus is similar to sirolimus in that they are both

immunosuppressive and antiproliferative agents. Everoli-

Fig. 4. This figure provides a comparison of the basic structure, molecular weigh

its analogues.

mus suppresses antigen-mediated T cell proliferation and

therefore has been used as an immunosuppressant. Ever-

olimus is a cytostatic agent, which acts by binding to the

intracellular receptor FKBP-12 resulting in late G1 cell

cycle arrest of vascular smooth cell proliferation. Unlike

sirolimus, everolimus has increased solubility in organic

solvents and has a two- to threefold lower affinity for the

receptor FKBP12. Structurally, the two compounds differ in

a side chain which for sirolimus is a hydrogen and for

everolimus is CH2(CH2)OH (Fig. 4). Preclinical data con-

ducted by Honda et al. [34] has demonstrated that ever-

olimus may be as effective as sirolimus in the prevention of

in-stent restenosis in a porcine coronary model.

The first clinical trial, to assess the safety and efficacy, of

everolimus-eluting stent was initiated at the Heart Center in

Siegburg, Germany by Dr. Eberhard Grube called ‘‘First

Use To Underscore Reduction in Restenosis with Ever-

olimus’’ (FUTURE I) [35]. The stent platform in this trial

is the Challenge stent, which is coated by a bioerodable

polymer-carrying everolimus. This was a prospective, ran-

domized and single-blinded trial of everolimus-eluting stent

in 40 patients with de novo coronary lesions, 2.75–4.0 mm

in diameter and <18 mm in length. Randomization was in a

2:1 ratio with 27 patients receiving a everolimus-eluting

stent and 13 patients receiving a bare metal stent. The pri-

mary endpoint was major adverse cardiac events at 30 days

and secondary endpoint is angiographic restenosis at

6 months. The preliminary results of FUTURE 1 were

presented by Dr. Eberhard Grube at the American College

of Cardiology, March 2003. Major adverse cardiac events at

t, physical properties, and approved clinical indication for rapamycin and

Page 8: Stent-based immunosuppressive therapies for the prevention of restenosis

M. Aggarwal et al. / Cardiovascular Radiation Medicine 4 (2003) 98–107 105

30 days did not differ significantly between the everolimus

group and control group. Angiographic study at 6-months

demonstrated late loss of 0.10 mm in the everolimus group

and 0.83 mm in the bare metal group. Although angio-

graphic restenosis (>50% diameter stenosis) was not ob-

served the everolimus group, this was not significantly

different from the 9.1% incidence of restenosis in the

control group. The results from the FUTURE I trial

demonstrated the feasibility and safety of this unique

bioerodable polymeric everolimus elluting stent. The FU-

TURE II is a multicenter prospective clinical trial, which

will consist of 200 patients to be randomized in a double-

blinded manner to an everolimus-eluting stent or a bare

metal stent. The primary end point of FUTURE II will be

angiographic late lumen loss at 6 months. Secondary

endpoints will be major adverse cardiac events at 1 and 6

months and 1 year.

6. Abt-578

ABT-578 is a synthetic analogue of sirolimus. The

molecular structure of ABT-578 contains a tetrazole ring

as compared to a hydroxyl group in sirolimus. Its mecha-

nism of action is similar to sirolimus in that ABT-578 binds

the intracellular receptor FKBP-12 and exerts an antiproli-

ferative effect via mTOR. In vitro data with ABT-578 has

demonstrated inhibition of growth-factor-mediated vascular

smooth muscle cell proliferation. Preclinical data has also

shown reduction in neointima formation at 28 days with

ABT-578 in the porcine coronary model. A Phase I clinical

trial with ABT-578-eluting phosphorylcholine-coated stent

is currently in-progress (ENDEAVOR I).

7. Myophenolic acid (MPA)

MPA (C7H20O6; MW 320.3) is an antibiotic, first derived

from cultures of the Penicillium species by Gosio in 1896

[36]. MPA has antineoplastic, antibacterial, antifungal, anti-

viral and excellent immunosuppressive properties. MPA is

the active metabolite of mycophenolic mofetil (MMF), an

FDA-approved drug, indicated for the prophylaxis of organ

rejection in patients receiving allogeneic renal, cardiac or

hepatic transplants. It is available for both oral (capsules,

tablets and oral suspension) and IV administration at rec-

ommended doses of 1–1.5 g b.i.d. (daily dose of 2–3 g) for

transplant patients.

MPA is a noncompetitive reversible inhibitor of IMPDH

(inosine monophosphate dehydrogenase). This NAD-

dependant enzyme is the rate-limiting enzyme in the de

novo pathway for purine biosynthesis and leads to depletion

of guanine nucleotides. This decrease in GTP results in

decreased DNA synthesis, allosteric feedback inhibition of

purine and pyrimidine biosynthesis, inhibition of glycosyl-

ation of adhesion molecules, and decreased cyclin depen-

dant kinase activity resulting in G0/G1 arrest [37].

MPA is known to exert antiproliferative activity on

lymphocytes, macrophages and smooth muscle cells [38].

In addition, depletion of guanine nucleotides inhibits trans-

fer of mannose and fructose to glycoproteins, preventing the

glycosylation of lymphocyte and monocyte glycoproteins

that are involved in adhesion to endothelial cells, thus,

inhibits recruitment of leukocytes into sites of inflammation

and graft rejection [39].

MPA is effective in the prevention of graft vascular

disease in rat models of aortic [40,41] and renal [42,43]

transplantation. Both the adventitial inflammatory compo-

nent and neointimal proliferation were inhibited by MPA in

the aortic transplant animals. In the renal transplant study,

allospecific IgM and IgG responses were absent in the

treated group. MPA, on its own and when given with

rapamycin was able to inhibit neointimal formation after

balloon injury [44]. In a study of heterotopic primate cardiac

xenografts [45], mycophenolate was more effective than

azathioprine when combined with cyclosporine and steroids

in preventing graft vascular disease. MPAwas also shown to

ameliorate the atherogenic potential of a high cholesterol diet

and to reduce macrophage and foam cell infiltration and

smooth muscle cell infiltration and proliferation [46].

MPA is incorporated in a proprietary polymer coating on

the Duraflex drug-eluting stent system developed by Avan-

tec Vascular (Sunnyvale, CA). The polymer employed by

Avantec has a long history in medical device industry

including blood-contacting and permanent implants. The

polymer coating is thin (V5 A), durable, and flexible and

can be modified to control the release rates of both hydro-

philic and hydrophobic drugs. The combination of the

coating technology with the inert nature of the polymer

results in no chemical interaction between the drug and the

polymer. The total amount of drug loaded on an 18 mm long

Duraflex stent is 300 Ag (3.3 Ag/mm2). The release of MPA

has been modified to provide a range of drug elution from

the stent for periods of 14 to 45 days.

In vitro studies have confirmed the broad therapeutic

window of MPA by measuring the antiproliferative effect

of MPA on human smooth muscle cells (SMCs) (prolifer-

ation assay) and the cytostatic effect of MPA on human

SMCs (viability assay). These studies suggest that MPA is

effective in inhibiting cell proliferation by a cytostatic

mechanism. MPA did not induce cell death even at con-

centrations 1000 times higher than the IC50 for inhibition of

SMC proliferation.

The IMPACT clinical trial (Inhibition with MPA of

Coronary Restenosis Trial) in February, 2002. This non-

randomized open label study was designed to evaluate the

safety and efficacy of the MPA-eluting Duraflex Stent

System in 150 patients with focal native de novo coronary

arterial lesions. The preliminary results of the IMPACT trial

presented during the EuroPCR in May 2003 indicated the

feasibility and safety of this drug-eluting stent but suggested

Page 9: Stent-based immunosuppressive therapies for the prevention of restenosis

M. Aggarwal et al. / Cardiovascular Radiation Medicine 4 (2003) 98–107106

minimal effect on late lumen loss. Further study is in-

progress to evaluate MPA-eluting stents with different

elution characteristics to increase early drug tissue concen-

tration as well as MPA in combination with other agents.

8. Conclusions

Stent based delivery of immunosuppressive agents has

established itself as a viable strategy for the reduction of in-

stent restenosis. The present data substantiates varying

degrees of clinical efficacy for stent-based immunosuppres-

sive therapies likely related to structural properties (MW,

solubility) and specific biologic effects based on mecha-

nism of action. Agents with a multiplicity of biologic

effects, such as sirolimus and other rapamycin analogs,

appear to produce the most potent suppression of neo-

intimal formation (90% inhibition) and thus the greatest

clinical benefit for reduction of restenosis. A ‘‘class effect’’

for these structurally and functionally divergent immuno-

suppressive compounds has not been observed with stent-

based delivery for the prevention of neointimal formation

and restenosis. For the sirolimus-eluting stent, the combi-

nation of efficacy and safety, with reduced restenosis and

absence of a pathobiological response, should make this a

core technology for the interventional cardiologist. As other

therapeutic agents and combination drug-eluting stents are

being explored, we must remember that all drugs and

carrier vehicles are not equivalent. The use of sirolimus

and other drug eluting stents in high-risk subset groups,

insulin-requiring diabetics, and in complex lesions, such as

at points of bifurcation requires additional study in larger

randomized clinical trials. In the coming years, emphasis

will shift to advancement of stent design for drug delivery

and ease of use as well as operator technique which will

demand vigilance at time of stent placement and deploy-

ment—goal to limit area of injury, avoid gaps in between

stents and perform optimal stent expansion. The stent–

carrier–drug composite system is a breakthrough medical

technology that will transform the principles and practice of

vascular medicine.

References

[1] Williams DO, Holubkov R, Yeh W, Bourassa MG, Al-Bassam M,

Block PC, Coady P, Cohen H, Cowley M, Dorros G, Faxon D,

Holmes DR, Jacobs A, Kelsey SF, King SB, Myler R, Slater J, Stanek

V, Vlachos HA, Detre KM. Percutaneous coronary intervention in the

current era compared with 1985–1986: the National Heart, Lung, and

Blood Institute Registries. Circulation 2000;102(24):2945–51.

[2] Mehran R, Dangas G, Abizaid AS, Mintz GS, Lansky AJ, Satler LF,

Pichard AD, Kent KM, Stone GW, Leon MB. Angiographic patterns

of in-stent restenosis: classification and implications for long-term

outcome. Circulation 1999;100(18):1872–8.

[3] Mehran R, Dangas G, Mintz GS, Waksman R, Hong MK, Abizaid A,

Abizaid AS, Kornowski R, Lansky AJ, Laird JR, Kent KM, Pichard

AD, Salter LF, Stone GW, Leon MD. In-stent restenosis: ‘‘The Great

Equalizer’’—disappointing clinical outcomes with ALL intervention-

al strategies. J Am Coll Cardiol 1999;33(Suppl. A):63A.

[4] Waksman R. Beta radiation to INHIBIT recurrance of in-stent reste-

nosis: clinical and angiographic results of the multicenter randomized

double blind study. Late breaking clinical trials at the American Heart

Association. New Orleans, LA, 2000.

[5] Kuntz RE. The beta-cath system trial. Late breaking clinical trials at

American College of Cardiology. Orlando, FL, 2001.

[6] Hoffmann R, Mintz GS, Dussaillant GR, Popma JJ, Pichard AD,

Satler LF, Kent KM, Griffin J, Leon MB. Patterns and mechanisms

of in-stent restenosis. A serial intravascular ultrasound study. Circu-

lation 1996;94(6):1247–54.

[7] Longenecker JP, Kilty LA, Johnson LK. Glucocorticoid influence

on growth of vascular wall cells in culture. J Cell Physiol 1982;

113:192–202.

[8] Voisard R, Seitzer U, Baur R, Dartsch PC, Osterhues H, Hoher M,

Hombach V. Corticosteroid agents inhibit proliferation of smooth

muscle cells from human atherosclerotic arteries in vitro. Int J Cardiol

1994;43(3):257–67.

[9] Cronstein BN, Kimmel SC, Levin RI, Martiniuk F, Weissmann G. A

mechanism for the antiinflammatory effects of corticosteroids: the

glucocorticoid receptor regulates leukocyte adhesion to endothelial

cells and expression of endothelial-leukocyte adhesion molecule 1

and intercellular adhesion molecule 1. Proc Natl Acad Sci U S A

1992;89(21):9991–5.

[10] Chervu A, Moore WS, Quinones-Baldrich WJ, Henderson T. Efficacy

of corticosteroids in suppression of intimal hyperplasia. J Vasc Surg

1989;10(2):129–34.

[11] Hollander W, Kramsch DM, Franzblau C. Suppression of athe-

romatous fibrous plaque formation by anti-proliferative and anti-

inflammatory drugs. Circ Res 1974;34:I-131–4 (supplement).

[12] Stone GW, Rutherford BD, McConahay DR, Johnson WL, Giorgi LV,

Ligon RW, Hartzler GO. A randomized trial of corticosteroids for the

prevention of restenosis in 102 patients undergoing repeat coronary

angioplasty. Catheter Cardiovasc Diagn 1989;18(4):227–31.

[13] Pepine CJ, Hirshfeld JW, Macdonald RG, Henderson MA, Bass TA,

Goldberg S, Savage MP, Vetrovec G, Cowley M, Taussig AS. A

controlled trial of corticosteroids to prevent restenosis after coronary

angioplasty. M-HEART Group. Circulation 1990;81(6): 1753–61.

[14] Lincoff AM, Furst JG, Ellis SG, Tuch RJ, Topol EJ. Sustained local

delivery of dexamethasone by a novel intravascular eluting stent to

prevent restenosis in the porcine coronary injury model. J Am Coll

Cardiol 1997;29(4):808–16.

[15] Strecker EP, Gabelmann A, Boos I, Lucas C, Xu Z, Haberstroh J,

Freudenberg N, Stricker H, Langer M, Betz E. Effect on intimal

hyperplasia of dexamethasone released from coated metal stents com-

pared with non-coated stents in canine femoral arteries. Cardiovasc

Intervent Radiol 1998;21(6):487–96.

[16] Ping QB, Yanming H, Wang L, Vermeire I, Verbeken E, Schacht E,

de Scheerder IK. Methylprednisolone coated stents decrease neo-

intimal hyperplasia in a porcine coronary model. J Am Coll Cardiol

2001;37(Suppl. 1):74A.

[17] Liu X, Huang Y, Scheerder ID. Study of antirestenosis with the Bio-

divysio dexamethasone eluting stent (STRIDE): a multicenter trial.

J Am Coll Cardiol 2002;39(Suppl. 1):15A.

[18] Meiser BM, Morris RE. Identification of a new pharmacologic action

for an old compound. Med Sci Res 1989;17:609–10.

[19] Gregory CR, Huie P, Billingham ME, Morris RE. Rapamycin inhibits

arterial intimal thickening caused by both alloimmune and mechanical

injury. Its effect on cellular, growth factor, and cytokine response in

injured vessels. Transplantation 1993;55(6):1409–18.

[20] Marx SO, Jayaraman T, Go LO, Marks AR. Rapamycin-FKBP inhib-

its cell cycle regulators of proliferation in vascular smooth muscle

cells. Circ Res 1995;76(3):412–7.

[21] Poon M, Marx SO, Gallo R, Badimon JJ, Taubman MB, Marks AR.

Rapamycin inhibits vascular smooth muscle cell migration. J Clin

Invest 1996;98(10):2277–83.

Page 10: Stent-based immunosuppressive therapies for the prevention of restenosis

M. Aggarwal et al. / Cardiovascular Radiation Medicine 4 (2003) 98–107 107

[22] Gallo R, Padurean A, Jayaraman T, Marx S, Roque M, Adelman S,

Chesebro J, Fallon J, Fuster V, Marks A, Badimon JJ. Inhibition

of intimal thickening after balloon angioplasty in porcine coronary

arteries by targeting regulators of the cell cycle. Circulation 1999;

99(16):2164–70.

[23] Suzuki T, Kopia G, Hayashi S, Bailey LR, Llanos G, Wilensky R,

Klugherz BD, Papandreou G, Narayan P, Leon MB, Yeung AC, Tio F,

Tsao PS, Falotico R, Carter AJ. Stent-based delivery of sirolimus

reduces neointimal formation in a porcine coronary model. Circula-

tion 2001;104(10):1188–93.

[24] Sousa JE, Costa MA, Abizaid A, Abizaid AS, Feres F, Pinto IM,

Seixas AC, Staico R, Mattos LA, Sousa AG, Falotico R, Jaeger J,

Popma JJ, Serruys PW. Lack of neointimal proliferation after implan-

tation of sirolimus-coated stents in human coronary arteries: a quan-

titative coronary angiography and three-dimensional intravascular

ultrasound study. Circulation 2001;103(2):192–5.

[25] Sousa JE, Costa MA, Abizaid AC, Rensing BJ, Abizaid AS, Tana-

jura LF, Kozuma K, Van Langenhove G, Sousa AG, Falotico R,

Jaeger J, Popma JJ, Serruys PW. Sustained suppression of neointi-

mal proliferation by sirolimus-eluting stents: one-year angiographic

and intravascular ultrasound follow-up. Circulation 2001;104(17):

2007–11.

[26] Morice MC, Serruys PW, Sousa JE, Fajadet J, Ban Hayashi E, Perin

M, Colombo A, Schuler G, Barragan P, Guagliumi G, Molnar F,

Falotico R. A randomized comparison of a sirolimus-eluting stent

with a standard stent for coronary revascularization. N Engl J Med

2002;346(23):1773–80.

[27] Leon MB. Sirius: SIRIUS 400. Late breaking clinical trials at Euro

PCR. Paris, France, 2002.

[28] Sousa JE, Abizaid AA, Sousa A, Costa MA, Evelyn R, Benno R, Mu-

zaffer D, Marinella C, Rodolfo S, Abizaid AA, Serruys PW, Tanajura

LF. Sirolimus-coated stents for the treatment of in-stent restenosis: the

first-in-man experience. Circulation 2001;104(17 [Suppl. II]):II-625.

[29] Degertekin M, Regar E, Tanabe K, Smits PC, van der Giessen WJ,

Carlier SG, de Feyter P, Vos J, Foley DP, Ligthart JMR, Popma JJ,

Serruys PW. Sirolimus-eluting stent for treatment of complex in-stent

restenosis. The first clinical experience. J Am Coll Cardiol 2003;41:

184–9.

[30] Shapiro R. The development of tacrolimus in renal transplantation.

Transplant Proc 2001;33:3158–60.

[31] Starzl TE, Todo S, Fung JJ, et al. Transplant Proc 22 [entire issue].

[32] Gummert JF, Ikonen T, Morris RE. Newer immunosuppressive drugs:

a review. J Am Soc Nephrol 1999;10(6):1366–80.

[33] Libby P, Schwartz D, Brogi E, Tanaka H, Clinton SK. A cascade

model for restenosis. A special case of atherosclerosis progression.

Circulation 1992;86(6 [Suppl. III]):III47–52.

[34] Honda H, Kar S, Honda T, Takizawa K, Meguro T, Fishbein MC,

Makkar R, Eigler N, Litvack F. Everolimus eluting stents significantly

inhibit neointimal hyperplasia in an experimental pig coronary model.

Am J Cardiol 2002;90(Suppl. 6A):73H.

[35] Grube E, Gerckens U, Buellesfeld L, Bootsveld A, Techen G, Staber-

ock M, Selbach G, Mueller R. First human experience using a new

everolimus stent coating: early findings of the FUTURE trial. Am J

Cardiol 2002;90(Suppl. 6A):71H.

[36] Gosio B. Ricerche Batteriologiche e chimiche sulle alterazioni del

mais. Revista di Igiene e Sanita Pubblica Ann 1896;7:825–68.

[37] Laurent AF, Dumont S, Poindron P, Muller CD. Mycophenolic acid

suppresses protein N-linked glycosylation in human monocytes and

their adhesion to endothelial cells and to some substrates. Exp Hem-

atol 1996;24(1):59–67.

[38] Allison AC, Eugui EM. Purine metabolism and immunosuppres-

sive effects of mycophenolate mofetil (MMF). Clin Transplant 1996;

10(1 Pt 2):77–84.

[39] Hauser IA, Johnson DR, Thevenod F, Goppelt-Strube M. Effect of

mycophenolic acid on TNF alpha-induced expression of cell adhesion

molecules in human venous endothelial cells in vitro. Br J Pharmacol

1997;122(7):1315–22.

[40] Morris RE, Hoyt EG, Murphy MP, Eugui EM, Allison AC. Myco-

phenolic acid morpholinoethylester (RS-61443) is a new immunosup-

pressant that prevents and halts heart allograft rejection by selective

inhibition of T- and B-cell purine synthesis. Transplant Proc 1990;

22(4):1659–62.

[41] Raisanen-Sokolowski A, Aho P, Myllarniemi M, Kallio E, Hayry P.

Inhibition of early chronic rejection in rat aortic allografts by myco-

phenolate mofetil (RS61443). Transplant Proc 1995;27(1):435.

[42] Azuma H, Binder J, Heemann U, Schmid C, Tullius SG, Tilney NL.

Effects of RS61443 on functional and morphological changes in

chronically rejecting rat kidney allografts. Transplantation 1995;

59(4):460–6.

[43] Azuma H, Binder J, Heemann U, Tullius SG, Tilney NL. Effect of

RS61443 on chronic rejection of rat kidney allografts. Transplant Proc

1995;27(1):436–7.

[44] Gregory CR, Huang X, Pratt RE, Dzau VJ, Shorthouse R, Billingham

ME, Morris RE. Treatment with rapamycin and mycophenolic

acid reduces arterial intimal thickening produced by mechanical in-

jury and allows endothelial replacement. Transplantation 1995;59(5):

655–61.

[45] O’Hair D, McManus RP, Komorowski R. Inhibition of chronic vas-

cular rejection in primate cardiac xenografts using mycophenolate

mofetil. Ann Thorac Surg 1994;58(5):1311–5.

[46] Romero F, Rodriguez-Iturbe B, Pons H, Parra G, Quiroz Y, Rincon J,

Gonzalez L. Mycophenolate mofetil treatment reduces cholesterol-

induced atherosclerosis in the rabbit. Atherosclerosis 2000;152(1):

127–33.