Next Article in Journal
Effect of Mo Content on Microstructure and Property of Low-Carbon Bainitic Steels
Next Article in Special Issue
Evaluation of Chill Cast Co-Cr Alloys for Biomedical Applications
Previous Article in Journal
Direct Metal Laser Sintering of Ti6Al4V for Biomedical Applications: Microstructure, Corrosion Properties, and Mechanical Treatment of Implants
Previous Article in Special Issue
Specific Yielding of Selective Laser-Melted Ti6Al4V Open-Porous Scaffolds as a Function of Unit Cell Design and Dimensions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Development of Coronary Artery Stents: From Bare-Metal to Bio-Resorbable Types

Department of Cardiology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
*
Author to whom correspondence should be addressed.
Metals 2016, 6(7), 168; https://doi.org/10.3390/met6070168
Submission received: 30 May 2016 / Revised: 26 June 2016 / Accepted: 14 July 2016 / Published: 20 July 2016
(This article belongs to the Special Issue Metallic Biomaterials)

Abstract

:
Coronary artery disease is the leading cause of death worldwide. Conventional balloon angioplasty is associated with high rates of complications such as coronary dissection and vessel recoil. The deployment of bare-metal stents (BMSs) can overcome these problems and achieve a better patency rate than simple balloon angioplasty. It has been shown that the stent design including structure platform, size, length, and strut thickness has a major influence on the clinical results. Even though angioplasty with BMS implantation is widely used in coronary interventions, the restenosis rate due to neointimal hyperplasia remains high. Therefore, drug-eluting stents (DESs) coated with anti-proliferative agents and polymers have been developed to reduce the restenosis rate and improve the clinical outcomes. Although the repeat revascularization rate of DESs is lower than that of BMSs, the long-term stent thrombosis rate is higher than for BMSs. Therefore, new and emerging generations of stents, in which, for example, thinner struts and bioresorbable polymers are used, are available for clinical use. However, there are only a limited number of clinical trials, in which these newer stents have been compared with BMSs and first- and second-generation DESs. The purpose of this review was to provide up-to-date information on the evolution of coronary artery stents from BMSs to DESs to bioresorbable stents (BRSs).

1. The Evolution of Coronary Stent Development

Coronary artery disease (CAD) is a major cause of mortality and morbidity. One of the treatment options for CAD is a percutaneous coronary intervention. Balloon angioplasty alone, which involves pushing the plaque to the walls of the artery, can achieve a larger luminal diameter in the acute phase. However, arterial recoil, dissection, and late neointimal hyperplasia can result in vessel restenosis. Therefore, bare metal stents (BMSs) made from metallic materials were developed to overcome the dissection and the restenosis complications caused by recoil. Although the restenosis rate can be reduced by stenting, the in-stent restenosis (ISR) rate is still high at around 20%–30% [1,2]. ISR is mostly caused by neointimal hyperplasia.
A reduction in restenosis was first achieved with the evolution of the stent platform, including thinner stent struts and new metal compounds. Furthermore, new drug delivery systems and polymer coatings have been developed to achieve better clinical outcomes. The first generation of drug-eluting stents (DESs) with a sirolimus or paclitaxel coating showed better clinical outcomes with lower rates of cardiac death, myocardial infarction, and target vessel revascularization than BMSs [3,4]. However, increased rates of stent thrombosis were reported with first-generation DESs, and second-generation DESs with thinner struts, different anti-proliferative drugs and polymers were subsequently developed to reduce the rate of long-term cardiac adverse events. More recently, bioresorbable material has been developed for use in stents. Even though the long-term outcomes are unknown, this material and new designs have changed the application of percutaneous coronary intervention.
In the present work, we review the evolution of coronary stents, from BMSs, through first-generation DESs, to today’s fully BRSs. Aspects covered in this review are materials, design, and clinical performance.

2. Stent Structure

A coronary stent is designed to maintain vessel patency. An ideal stent should have good biocompatibility, flexibility and deliverability, low thrombogenesis, strong radial force to maintain a large vessel diameter, good radio-opacity under fluoroscopy, and low rates of neointimal hyperplasia and stent thrombosis during long-term follow-up.
A classic stent is composed of three components: a metallic platform, polymer coating, and anti-proliferative drug. BMSs are constructed from metallic alloys without drug elution, including many different platforms such as stainless steel, platinum-chrome, tantalum, and cobalt-chrome. Stainless steel (316 L) is a mixture of 95% iron and 5% nickel, and is poorly visible under fluoroscopy. Tantalum alloys have poor radial force and easily recoil. Cobalt-chrome alloys have better radial strength and radio-opacity, and most stents are currently made from chrome with cobalt or platinum struts. Platinum-chrome alloys are twice as dense as cobalt-chrome alloys, and so have better radio-opacity under fluoroscopy. Alloys other than those containing stainless steel can allow for thinner struts while maintaining radial force. The first-generation DESs had thick struts of around 130–140 μm, while the next generation had thinner struts of around 70–90 μm, resulting in a decrease in both the rates of angiographic and clinical restenosis [5]. The coating of the metallic surface, including chromium oxide or gold, can reduce biological interactions with blood, however, a gold surface has been shown to result in a high incidence of restenosis. Except for balloon-expandable stents, most self-expandable stents are made from nitinol, which can maintain its initial shape within the vessel.
Even with the same material, different configurations can be used to create different designs. Stent designs include wire coils, slotted tubes, and a modular design. Wire coils are formed using 0.15 mm circular metallic wire to create loops and folds for expansion. However, few connections between struts results in good flexibility but poor radial force, and coil-type stents are thus no longer available. Slotted tubular stents were designed to replace wire coils stents to increase radial strength, and they include some articulation between the stent segments. The drawback of this design is poor flexibility due to the rigid structure, although recent improvements in design using laser cutting have resulted in improved flexibility without compromising strength. Another issue in stent design is the use of closed or open cells in multicellular stents. An open cell design allows for better flexibility and deliverability, side branch access and tolerability on bending. In comparison, a closed cell design can provide better vessel coverage and radial strength but less flexibility. Modular stents can overcome the problems with flexibility in slotted tube stents. They are comprised of multiple repeat modules joined together to construct a stent tube, which can provide good flexibility and side branch access.
The cross-sectional shape of a stent is another factor in its design. Some stents have round struts with no edge or square struts. Round struts can reduce turbulent blood flow and avoid cells’ disruption. In addition, different sizes and lengths of the stent can also affect the clinical outcomes. A large diameter involves a large vessel area and less late loss, whereas a longer length of stent increases the rate of restenosis in BMSs but not in DESs [6]. The stent coatings are designed to reduce thrombotic events and restenosis, and include heparin-based, phosphorylcholine (PC), and carbon.
Another important component of a stent is the polymer, which is used as a carrier for anti-proliferative drug release. Polymers have been reported to result in an inflammatory effect leading to thrombosis [7]. To overcome this problem, newer stents are polymer-free or use bioresorbable polymers. As a polymer degrades, endothelial healing improves, and so polymer-free or bioresorbable stent designs may prevent the early complications of BMSs and improve the later complications of DESs. This will theoretically reduce the rate of stent thrombosis, however the long-term outcomes need to be verified in large randomized controlled trials.

3. Bare Metal Stents

Coronary angiography and balloon angioplasty were introduced in the late 1970s. Initially, plain balloon angioplasty without stent implantation showed high rates of arterial recoil and vessel dissection. The first coronary stent was implanted in 1986. Coronary stents can seal dissected tissue and avoid elastic recoil, and they have been shown to result in better outcomes than plain balloon angioplasty [1,2]. Therefore, coronary stents have become the most common therapeutic intervention for CAD. However, BMSs have a high rate of ISR due to neointimal hyperplasia with smooth muscle migration and proliferation [8]. A previous trial reported an ISR rate of 10%–20% during six months of follow-up which resulted in myocardial infarction and angina requiring revascularization [9].
Initial stents made from stainless steel have thick struts. Because of poor flexibility and high ISR rate, stents with thin struts were developed [5,10]. The stainless steel was replaced by cobalt, chromium, or other elements to maintain the strength and radio-opacity. Cobalt-chromium alloys are used in Multi-Link Vision L605 (Abbot Vascular, CA, USA), Coroflex (B. Braun, Berlin, Germany), and Driver MP35N (Medtronic, Santa Rosa, CA, USA) stents [11,12]. Other compounds, including a platinum-chromium alloy in Omega (Boston Scientific, Natick, MA, USA) stents and a cobalt alloy in Integrity (Medtronic, Santa Rosa, CA, USA) stents were developed. These compounds allow for thinner struts and have better radio-opacity, deliverability, and conformability, however the ISR rate is still high at around 15% after follow-up of 6 months [11,12].
Several inorganic coatings on the stent surface have been developed to minimize metal ion release, including diamond-like carbon, iridium oxide, titanium-nitride-oxide, and silicon carbide. However, clinical trials have not shown any benefits in the ISR rate with these coatings [13,14,15,16]. Therefore, the development of a drug-eluting polymer coating has become an important area of research to overcome this problem. Despite the application of new anti-proliferative drug-eluting technology, the stent platform and coating still play an important role in stent design, even though polymer-free stents and those using bioresorbable polymers are used currently.

4. Drug-Eluting Stents

The application of anti-proliferative agents was a major breakthrough in coronary artery stent design. The major goal of a DES is to reduce ISR by inhibiting neointimal hyperplasia. Several factors affect the drug elution from polymers, and how best to release the drug and the duration of drug elution are the most important issues in DES design.

4.1. First-Generation DESs

The structure of the first generation of DESs was based on a stainless steel platform coated with a drug-eluting polymer. Two widely used DESs were the Cypher (Cordis Corporation, CA, USA) and Taxus (Boston Scientific, Natick, MA, USA) stents first introduced in 2003, both of which were made of stainless steel and had a stent strut thickness of about 135 μm. The Cypher stent had a Parylene tie-layer and polyethylene-co-vinyl acetate (PEVA) and poly-n-butyl methacrylate (PBMA) polymer with a sirolimus coating. Sirolimus is an immunosuppressant which can inhibit cell proliferation in the G1 to S phase of the cell cycle. The Taxus Express and Taxus Liberte (Boston Scientific, Natick, MA, USA) stents had a poly(styrene-b-isobutylene-b-styrene) (SIBS) polymer with a paclitaxel coating [17]. Paclitaxel is approved for the treatment of ovarian cancer via inhibiting microtubules for mitosis. Both sirolimus and paclitaxel have anti-proliferative effects which can inhibit smooth muscle migration and endothelial healing. The first-generation DESs were reported to have better efficacy with lower rates of ISR and target vessel revascularization than BMSs in a meta-analysis study, with a reduction in the rate of target vessel revascularization by 60%–70% [18]. Other trials have reported that sirolimus-eluting stents are better than paclitaxel-eluting stents with regards to target lesion revascularization, stent thrombosis and ISR [19,20,21].
However, after DES implantation, increased rates of myocardial infarction and cardiac death caused by stent thrombosis have been reported during long-term follow-up [22,23,24]. Stent thrombosis can cause abrupt vessel closure resulting in serious cardiovascular events. The Academic Research Consortium (ARC) reported that the majority of cases of stent thrombosis occur late or very late, with a very late rate of around 0.2%–0.5% per year, which is higher than for BMSs [25]. The high rate of late and very late stent thrombosis is caused by a long duration of drug elution, which can delay endothelial healing and prolong metallic structure exposure to blood vessel [26]. Interestingly, anti-proliferative agents have been reported to reduce the rate of ISR but increase the rate of stent thrombosis. Therefore, how best to balance efficacy and safety is an important issue.

4.2. Second Generation DESs

Second generation DESs were developed to overcome the problem of late and very late stent thrombosis by accelerating vessel healing. These DESs showed improvements in strut thickness, deliverability, and flexibility. The platform was changed to cobalt-chromium or platinum-chromium which allowed for a reduction in the strut thickness. These new stents used more biocompatible polymers such as PC, polylactic acid (PLA), poly(vinylidencefluoride-cohexafluoropropenen) (PVDF-HFP), and polyvinylpyrrolidone (PVP). New coating drugs were also used in second generation DESs, including zotarolimus, everolimus, and novolimus, which were shown to result in good endothelial coverage in an animal study [27].
Zotarolimus is used in the Endeavor and Endeavor Resolute (Medtronic Vascular, CA, USA) stents. The Endeavor stent consists of a cobalt-chromium platform with thinner struts and a PC polymer which has been shown to have better biocompatibility and anti-inflammatory effects, and to provide faster drug elution with earlier vessel healing. Randomized controlled trials have reported that this stent has better results in terms of ISR and repeat revascularization than a BMS [28,29]. The ENDEAVOR III trial reported that a zotarolimus-eluting stent was associated with lower major cardiac events during a five-year follow-up compared to a sirolimus-eluting stent (Cypher, CA, USA), but an increased rate of repeat revascularization was reported in a meta-analysis [29,30]. Moreover, a zotarolimus-eluting stent was associated with a lower rate of cardiac adverse events than a sirolimus-eluting stent in very late stent thrombosis after three years of follow-up [31]. However, Camenzind et al. reported no difference in clinical outcomes at one year between Endeavor and Taxus stents, although the Endeavor stent had a significantly lower rate of stent thrombosis between one and five years [32]. The ZEST trial also showed that the Endeavor stent had better outcomes than a sirolimus-eluting stent, but similar results to a paclitaxel-eluting stent after one year of follow-up [33]. Further generations of the Resolute stent used the same cobalt-chromium platform but a polymer called biolinx consisting of a mixture of a hydrophilic component on the endoluminal surface, and a hydrophobic component on the metal surface. This polymer, which was designed to prevent thrombotic events, can release 80% of the drug within 60 days compared to a typical range of 10 to 30 days in other stents.
Everolimus is a cell cycle inhibitor. Its mechanism of action is the same as sirolimus by inhibiting smooth muscle cell proliferation. Currently available everolimus stents include the Promus (Boston Scientific, Natick, MA, USA) and Xience (Abbott Vascular, CA, USA) stents, both of which use a cobalt-chromium platform. In comparison, the Promus Element stent (Boston Scientific, Natick, MA, USA) uses a platinum chromium alloy platform with thinner struts (81 μm) than the Taxus Liberte stent (96 μm) and better deliverability. Everolimus-eluting stents have also been reported to have lower rates of ISR and repeat revascularization due to less neointimal hyperplasia than BMSs [34,35]. Compared to paclitaxel-eluting stents, everolimus-eluting stents have been reported to have better clinical outcomes at one year of follow-up [36,37], and similar clinical outcomes to sirolimus-eluting stents [35,38,39]. Another trial reported that everolimus-eluting stents were associated with a low rate of major cardiac adverse events after five years of follow-up, due to a low rate of very late stent thrombosis [38]. Other clinical trials have reported similar results between Resolute and Xience V (Abbott Vascular, CA, USA) stents [40,41]. In summary, second generation DESs with everolimus or zotarolimus have been reported to have lower rates of very late stent thrombosis in multiple randomized controlled trials.
Novolimus is another novel drug. It is used in the DESyne (Elixir Medical, Sunnyvale, CA, USA) stent, and it is a mammalian target of rapamycin inhibitor. The thickness of the polymer coating is thinner (3 μm) than other (5–8 μm) stents, and a small randomized controlled trial reported no differences in results between novolimus-eluting and zotarolimus-eluting stents [42].
Even though BMSs are not as good as DESs, they are used in some situations. For example, the shorter duration of dual anti-platelet therapy is required with the use of BMSs due to rapid endothelialization after stenting, and they can be used in patients scheduled to undergo surgery and in those who cannot tolerate long-term dual anti-platelet therapy. BMSs have also been shown to have similar clinical outcomes in large coronary arteries (>3 mm) in terms of cardiac death and myocardial infarction compared to DESs [35].

5. Polymer-Free DESs

Theoretically, the polymer plays an important role in vessel healing. Polymer-free stents provide the benefits of vascular healing and less inflammation [43], and this may be beneficial in decreasing the rate of stent thrombosis during long-term follow-up. Therefore, it is possible that a shorter duration of dual anti-platelet therapy is safe with the use of polymer-free stents [44]. However, the problem is how to deliver anti-proliferative agents without a polymer. The function of the polymer is not only as a drug carrier, but also in controlling release of the drug. To develop polymer-free DESs, new technology was needed to allow for drug loading and released from the stent surface. Initially, direct loading of paclitaxel onto stainless steel failed to show better clinical outcomes than BMSs [45]. However, several stents were subsequently developed to achieve drug delivery without the use of a polymer. The stent surface was first modified to be porous, and then the drug was directly coated onto the stent surface by chemical bonding. Initially, nanopores (5–15 nm) were created on an aluminum stent surface. The drug was then loaded onto these pores and released after stent implantation. However, the rate of drug release was difficult to control from the nanopores.
The Yukon stent (Translumina, Hechingen, Germany) has a microporous stainless steel surface which can be coated with sirolimus. The stent should be used immediately after coating, and the drug is mostly eluted within seven days. An optical coherence tomography study of the stent coverage of the endothelium showed this stent to be better than sirolimus-eluting stents after three months [46]. Compared to paclitaxel-eluting stents, no differences in clinical outcomes were reported at five years of follow-up in the study by King et al. [47]. The ISAR-TEST 5 trial showed that this strut had non-inferior results at one year compared to a second-generation DES (zotarolimus-eluting stent) [48].
The drug can also be carried by nanoparticles in a matrix compound on a platform as used in other stents. These nanoparticles can facilitate penetration of the drug deeper into vessel walls and rapidly elute the drug. The Cre8 stent (CID Vascular, Saluggia, Italy) is a nanoparticle polymer-free stent that has an ultra-thin passive carbon coating on cobalt-chrome alloy. The anti-proliferative drug used is amphilimus, which was produced from sirolimus in a long-chain fatty acid mixture and is loaded into an abluminal reservoir. Compared to paclitaxel-eluting stents, Cre8 has shown a lower rate of in-stent late lumen loss but no difference in clinical end points [49]. The BioFreedom (Biosensors, Morges, Switzerland) stent has a stainless steel platform with a microstructure abluminal surface. Biolimus A9 is attached to the abluminal surface without a polymer. After stent implantation, one month of dual anti-platelet therapy in patients at high-risk of bleeding has shown better efficacy and safety than BMSs [44].
The Amazonia Pax (Minvasys, Gennevillieres, France) stent has cobalt-chromium platform with paclitaxel applied as microdrops on the abluminal surface by crystallization, and the drug can be released within 30 days. The VESTAsync (MIV Therapeutics, Atlanta, GA, USA) stent has a stainless steel platform, and a hydroxyapatite surface coated with sirolimus on microporous pores. The hydroxyapatite dissolves gradually within one year. The OPTIMA (CID, Saluggia, Italy) stent has a stainless steel platform without a polymer, and tacrolimus coated into grooves as a reservoir on the stent abluminal surface.
Few randomized controlled trials have evaluated the performance of polymer-free DESs, and relevant trials on the long-term efficacy and safety compared to second-generation DESs are needed. Regarding stent technology, modification of the stent surface may influence the stent structure. The drug reservoir and release are other issues. The development of new technology and bio-materials is still needed.

6. Partially-Bioresorbable Stents

In these stents, the drug is embedded in a coating made of a bioresorbable polymer and the coating is deposited on a durable material, usually a metal. According to previous studies, stent thrombosis remains a concern after stent implantation with both BMSs and DESs. The risk factors include stent malapposition, inappropriate stent size, inadequate anti-platelet agents, delayed vessel healing, and chronic inflammation. The polymer plays an important role in the delay of vessel healing and inflammation [50,51]. No endothelial coverage of the stent surface will result in a higher rate of stent thrombosis. At least six months of dual anti-platelet therapy is currently recommended after DES implantation to decrease stent thrombosis. However, endothelial coverage can still be incomplete after six months. Even if second-generation DESs can improve vessel healing and reduce stent thrombosis, the residual polymer can still be a clinical concern. To avoid this effect, bioresorbable polymers were developed. The optimal degradation of these polymer, biocompatibility, and formulation are the most important factors in the design of bioresorbable polymers, as well as the long-term safety of the degradable compound. Several polymers are currently in use for drug elution, such as poly(d,l)lactic acid (PDLLA), poly(lactide-co-glycolide) (PLGA), and poly-l-lactic acid (PLLA). These bioresorbable polymers had better biocompatibility including endothelial function, smooth muscle cell growth, and thrombogenicity than permanent polymers such as PEVA and PBMA [52,53].
The stents that use PDLLA as the polymer include BioMatrix (Biosensor, Morges, Switzerland), Nobori (Terumo, Tokyo, Japan), Yukon Choice PC (Translumina GmbH, Hechingen, Germany), and Axxess (Biosensors, Morges, Switzerland). The stents which use PLLA as the polymer include Orsiro (Biotronik, Berlin, Germany) and DESyne BD (Elixir, CA, USA). The stents that use PLGA as the polymer include Synergy (Boston Scientific, Natick, MA, USA) and MiStent (Micell Technologies, Durham, USA). The stents that use multiple polymers include Ultimaster (Terumo, Tokyo, Japan) with mixture of PDLLA and poly(l-lactide co-e-caprolactone) (PCL), BioMime (Meril Life Sciences, Gujarat, India) with mixture of PLLA and PLGA, Combo (OrbusNeich Medical, FL, USA) with PLLA and PLGA, Infinium (Sahajanand, Gujarat, India) and Supralimus Core (Sahajanand, Gujarat, India) with mixture of PLLA, PCL, PVP, and PLGA (Summary in Table 1). According to the compound used, these polymers will biodegrade within different periods of time.
Biolimus-eluting stents have showed better clinical outcomes than BMSs in patients with ST-elevation myocardial infarction [54]. In addition, they have reported to have significantly lower rates of very late stent thrombosis compared to sirolimus-eluting durable polymer stents [55,56], and non-inferior clinical outcomes to Synergy (everolimus-eluting), Nobori (biolimus-eluting), Ultimaster (sirolimus-eluting), and Orsiro (sirolimus-eluting) stents [57,58,59,60,61,62,63]. The Nobori stent uses the same anti-proliferative drug and stainless steel platform, but an ultra-thin non-degradable Parylene coating between the stent and bioresorbable polymer. Moreover, the SORT OUT V trial compared the Nobori stent and first generation sirolimus-eluting stents, and reported similar results at one year [64]. In addition, compared to a zotarolimus-eluting stent with a durable polymer, a bioresorbable polymer-coated biolimus-eluting stent showed similar results at 1 year of follow-up (SORT OUT VI) [65]. The Orsiro stent has an ultrathin strut (60–80 μm) with a cobalt-chromium alloy and an amorphous hydrogen-rich silicon-carbide layer, which can reduce iron release. Sirolimus is loaded onto the PLLA polymer and can be released within 12–14 weeks, with the polymer degrading within 12–24 months. The SORT OUT VII trial comparing the efficacy and safety between Orsiro and Nobori stents is currently ongoing.
Another stent uses laser drilling technology to create drug-filled holes on the stent surface. The CoStar (Conor MedSystems, Palo Alto, CA, USA) stent is made from a cobalt-chrome alloy and a bioresorbable PLGA polymer with paclitaxel. These holes can avoid long-term exposure of the polymer to the vessel wall. The COSTAR II trial reported that the CoStar stent was not non-inferior to the Taxus DES regarding clinical or angiographic outcomes [66].
In summary, partially bioresorbable polymer DESs have lower rates of very late stent thrombosis than first generation DESs, with similar results to second generation DESs. Partially bioresorbable polymer DESs have also been shown to have non-inferior results compared to second generation DESs at one year; in one meta-analysis including 11 randomized clinical trials the biodegradable polymer stents didn’t show advantageous outcomes of stent thrombosis, target lesion revascularization, myocardial infarction, and cardiac death than permanent polymer stents [67]. However, further studies are needed regarding the long-term outcomes.

7. Fully Bioresorbable Stents (BRSs)

Using bioresorbable material to construct a stent is an active area of research. Bioresorbable stents are materials that can be biodegraded over time. The radial force and mechanical support are provided initially until the structure degrades. The rationale of using a bioresorbable material is to avoid the long-term development of neoatherosclerosis, stent fracture, and stent thrombosis. It can also connect native vessels to graft vessels if bypass surgery is needed. Bioresorbable material can also restore normal endothelial function in the vessels because metallic stents are not used. This new kind of stent may decrease the risk of stent thrombosis during long-term follow-up [68].
The stents are made from polymeric material, including PLLA, salicylic acid, or poly-tyrosine-derived polycarbonate and metallic material, including magnesium or zinc (Summary in Table 2). PLLA has better biocompatibility but thick struts were needed to provide mechanical strength. Thick struts may result in incomplete expansion and reduce lumen diameter. Metallic stents had better mechanical strength with thinner struts. However, magnesium had higher degradation rate and raise a problem of toxicity. Metallic Zinc is a new element for future stent design and provides low potential for restenosis and toxicity [69]. An initial clinical trial of a scaffold with magnesium reported a high restenosis rate due to the unstable release of the drug, however the later BIOSOLVE-1 trial reported better results [70,71]. The DREAMS (Biotronik, Bülach, Switzerland) scaffold uses an absorbable magnesium alloy with paclitaxel. The absorption process is complete after 9–12 months. There is a 1 μm absorbable PLGA polymer on the scaffold surface to carry the anti-proliferative drug. The DREAMS 2G (Biotronik, Bülach, Switzerland) scaffold is a next generation scaffold that uses sirolimus elution with a PLLA polymer. The structure is also made from an absorbable magnesium alloy, with fluoroscopic markers at both distal and proximal ends. The magnesium alloy is metabolized to hydrated magnesium oxide and magnesium phosphate which can be absorbed by the body. The non-randomized BIOSOLVE-II trial showed its safety and efficacy at six months of follow-up [72].
The radial force of a BRS is weaker than a DES, so the recoil can be a problem because of the rapid absorption [73]. To overcome this problem, the stent design requires thick struts to maintain the radial strength. The DESolve (Elixir Medical, CA, USA) stent was made from PLLA. The eluting drug as myolimus is resorbed within 1 year, and the radial strength can be maintained for 3–4 months. A small trial showed its safety and efficacy at one year of follow-up [74]. Newer DESolve stent decreased strut thickness to 100 μm and used the drug of novolimus. Another absorbable material is PLLA known as the “Bioresorbable Vascular Scaffold” (BVS; Abbot Vascular, CA, USA) with everolimus. The thickness of the scaffold is 150 μm. An initial trial reported a low rate of major cardiac adverse events at four years of follow-up [68,75]. Compared to everolimus-eluting stents, the BVS was reported to have non-inferiority results at one year of follow-up but a higher rate of subacute thrombosis, which may be due to greater strut thickness [76,77]. One meta-analysis also demonstrated similar result [78].
A fully BRS is another choice of coronary artery intervention, and its use is increasing rapidly. However, further randomized controlled trials are still needed to compare the long-term clinical benefits with new DESs. Investigation of a new zinc stent is another way to provide benefits for mechanical strength and restenosis [69].

8. Conclusions

Coronary angioplasty and stent implantation are treatment options for CAD. The high restenosis rate of BMSs can be overcome by the development of new DESs. Paclitaxel and the “-limus” family can inhibit neointimal growth. However, first generation DESs have the concerns of long-term safety due to a high incidence of late and very late stent thrombosis. Second generation DESs with a thin structure, different polymers and new anti-proliferative drugs that can be quickly eluted have been shown to be effective and safe. Everolimus- or zotarolimus-eluting stents were shown to decrease the rate of very late stent thrombosis in many randomized controlled trials. Furthermore, stents with bioresorbable polymers or polymer-free structures may also improve long-term outcomes. However, further studies comparing these designs with second generation DESs are needed.
Fully BRSs are the latest innovation, and recent data show non-inferior results to second generation DESs at one year of follow-up. In the future, the long-term outcomes of CAD may be improved by different struts or materials.

Acknowledgments

Chang Gung Memorial Hospital, Research project, I-Chang Hsieh: CORPG3C0162.

Author Contributions

Conceived and designed the review: Ming-Yun Ho, Chun-Chi Chen, I-Chang Hsieh. Collecting data: Chao-Yung Wang, Shang-Hung Chang. Contributed materials/table/reference: Ming-Jer Hsieh, Cheng-Hung Lee. Wrote the paper: Ming-Yun Ho, Victor Chien-Chia Wu, I-Chang Hsieh.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Serruys, P.W.; de Jaegere, P.; Kiemeneij, F.; Macaya, C.; Rutsch, W.; Heyndrickx, G.; Emanuelsson, H.; Marco, J.; Legrand, V.; Materne, P.; et al. A comparison of balloon-expandable-stent implantation with balloon angioplasty in patients with coronary artery disease. N. Engl. J. Med. 1994, 331, 489–495. [Google Scholar] [CrossRef] [PubMed]
  2. Fischman, D.L.; Leon, M.B.; Baim, D.S.; Schatz, R.A.; Savage, M.P.; Penn, I.; Detre, K.; Veltri, L.; Ricci, D.; Nobuyoshi, M.; et al. A randomized comparison of coronary-stent placement and balloon angioplasty in the treatment of coronary artery disease. N. Engl. J. Med. 1994, 331, 496–501. [Google Scholar] [CrossRef] [PubMed]
  3. Stone, G.W.; Ellis, S.G.; Cox, D.A.; Hermiller, J.; O’Shaughnessy, C.; Mann, J.T.; Turco, M.; Caputo, R.; Bergin, P.; Greenberg, J.; et al. A polymer-based, paclitaxel-eluting stent in patients with coronary artery disease. N. Engl. J. Med. 2004, 350, 221–231. [Google Scholar] [CrossRef] [PubMed]
  4. Moses, J.W.; Leon, M.B.; Popma, J.J.; Fitzgerald, P.J.; Holmes, D.R.; O’Shaughnessy, C.; Caputo, R.P.; Kereiakes, D.J.; Williams, D.O.; Teirstein, P.S.; et al. Sirolimus-eluting stents versus standard stents in patients with stenosis in a native coronary artery. N. Engl. J. Med. 2003, 349, 1315–1323. [Google Scholar] [CrossRef] [PubMed]
  5. Pache, J.; Kastrati, A.; Mehilli, J.; Schuhlen, H.; Dotzer, F.; Hausleiter, J.; Fleckenstein, M.; Neumann, F.J.; Sattelberger, U.; Schmitt, C.; et al. Intracoronary stenting and angiographic results: Strut thickness effect on restenosis outcome (ISAR-STEREO-2) trial. J. Am. Coll. Cardiol. 2003, 41, 1283–1288. [Google Scholar] [CrossRef]
  6. Chang, S.H.; Chen, C.C.; Hsieh, M.J.; Wang, C.Y.; Lee, C.H.; Hsieh, I.C. Lesion length impacts long term outcomes of drug-eluting stents and bare metal stents differently. PLoS ONE 2013, 8, e53207. [Google Scholar] [CrossRef] [PubMed]
  7. Van der Giessen, W.J.; Lincoff, A.M.; Schwartz, R.S.; van Beusekom, H.M.; Serruys, P.W.; Holmes, D.R., Jr.; Ellis, S.G.; Topol, E.J. Marked inflammatory sequelae to implantation of biodegradable and nonbiodegradable polymers in porcine coronary arteries. Circulation 1996, 94, 1690–1697. [Google Scholar] [CrossRef] [PubMed]
  8. Hoffmann, R.; Mintz, G.S.; Dussaillant, G.R.; Popma, J.J.; Pichard, A.D.; Satler, L.F.; Kent, K.M.; Griffin, J.; Leon, M.B. Patterns and mechanisms of in-stent restenosis. A serial intravascular ultrasound study. Circulation 1996, 94, 1247–1254. [Google Scholar] [CrossRef] [PubMed]
  9. Chen, M.S.; John, J.M.; Chew, D.P.; Lee, D.S.; Ellis, S.G.; Bhatt, D.L. Bare metal stent restenosis is not a benign clinical entity. Am. Heart J. 2006, 151, 1260–1264. [Google Scholar] [CrossRef] [PubMed]
  10. Briguori, C.; Sarais, C.; Pagnotta, P.; Liistro, F.; Montorfano, M.; Chieffo, A.; Sgura, F.; Corvaja, N.; Albiero, R.; Stankovic, G.; et al. In-stent restenosis in small coronary arteries: Impact of strut thickness. J. Am. Coll. Cardiol. 2002, 40, 403–409. [Google Scholar] [CrossRef]
  11. Kereiakes, D.J.; Cox, D.A.; Hermiller, J.B.; Midei, M.G.; Bachinsky, W.B.; Nukta, E.D.; Leon, M.B.; Fink, S.; Marin, L.; Lansky, A.J.; et al. Usefulness of a cobalt chromium coronary stent alloy. Am. J. Cardiol. 2003, 92, 463–466. [Google Scholar] [CrossRef]
  12. Sketch, M.H., Jr.; Ball, M.; Rutherford, B.; Popma, J.J.; Russell, C.; Kereiakes, D.J.; Driver, I. Evaluation of the medtronic (driver) cobalt-chromium alloy coronary stent system. Am. J. Cardiol. 2005, 95, 8–12. [Google Scholar] [CrossRef] [PubMed]
  13. Kim, Y.H.; Lee, C.W.; Hong, M.K.; Park, S.W.; Tahk, S.J.; Yang, J.Y.; Saito, S.; Santoso, T.; Quan, L.; Ge, J.; et al. Randomized comparison of carbon ion-implanted stent versus bare metal stent in coronary artery disease: The Asian Pacific Multicenter Arthos Stent Study (PASS) trial. Am. Heart J. 2005, 149, 336–341. [Google Scholar] [CrossRef] [PubMed]
  14. Meireles, G.C.; de Abreu, L.M.; Forte, A.A.; Sumita, M.K.; Sumita, J.H.; Jdel, C.A. Randomized comparative study of diamond-like carbon coated stainless steel stent versus uncoated stent implantation in patients with coronary artery disease. Arq. Bras. Cardiol. 2007, 88, 390–395. [Google Scholar] [CrossRef] [PubMed]
  15. Unverdorben, M.; Sippel, B.; Degenhardt, R.; Sattler, K.; Fries, R.; Abt, B.; Wagner, E.; Koehler, H.; Daemgen, G.; Scholz, M.; et al. Comparison of a silicon carbide-coated stent versus a noncoated stent in human beings: The Tenax versus Nir Stent Study’s long-term outcome. Am. Heart J. 2003, 145, e17. [Google Scholar] [CrossRef] [PubMed]
  16. Windecker, S.; Simon, R.; Lins, M.; Klauss, V.; Eberli, F.R.; Roffi, M.; Pedrazzini, G.; Moccetti, T.; Wenaweser, P.; Togni, M.; et al. Randomized comparison of a titanium-nitride-oxide-coated stent with a stainless steel stent for coronary revascularization: The TiNOX trial. Circulation 2005, 111, 2617–2622. [Google Scholar] [CrossRef] [PubMed]
  17. Turco, M.A.; Ormiston, J.A.; Popma, J.J.; Mandinov, L.; O’Shaughnessy, C.D.; Mann, T.; McGarry, T.F.; Wu, C.J.; Chan, C.; Webster, M.W.; et al. Polymer-based, paclitaxel-eluting TAXUS Liberte stent in de novo lesions: The pivotal TAXUS ATLAS trial. J. Am. Coll. Cardiol. 2007, 49, 1676–1683. [Google Scholar] [CrossRef] [PubMed]
  18. Stettler, C.; Wandel, S.; Allemann, S.; Kastrati, A.; Morice, M.C.; Schomig, A.; Pfisterer, M.E.; Stone, G.W.; Leon, M.B.; de Lezo, J.S.; et al. Outcomes associated with drug-eluting and bare-metal stents: A collaborative network meta-analysis. Lancet 2007, 370, 937–948. [Google Scholar] [CrossRef]
  19. Windecker, S.; Remondino, A.; Eberli, F.R.; Juni, P.; Raber, L.; Wenaweser, P.; Togni, M.; Billinger, M.; Tuller, D.; Seiler, C.; et al. Sirolimus-eluting and paclitaxel-eluting stents for coronary revascularization. N. Engl. J. Med. 2005, 353, 653–662. [Google Scholar] [CrossRef] [PubMed]
  20. Kastrati, A.; Dibra, A.; Eberle, S.; Mehilli, J.; de Lezo, J.S.; Goy, J.J.; Ulm, K.; Schomig, A. Sirolimus-eluting stents vs. paclitaxel-eluting stents in patients with coronary artery disease: Meta-analysis of randomized trials. JAMA 2005, 294, 819–825. [Google Scholar] [CrossRef] [PubMed]
  21. Schomig, A.; Dibra, A.; Windecker, S.; Mehilli, J.; de Lezo, J.S.; Kaiser, C.; Park, S.J.; Goy, J.J.; Lee, J.H.; di Lorenzo, E.; et al. A meta-analysis of 16 randomized trials of sirolimus-eluting stents versus paclitaxel-eluting stents in patients with coronary artery disease. J. Am. Coll. Cardiol. 2007, 50, 1373–1380. [Google Scholar] [CrossRef] [PubMed]
  22. Camenzind, E.; Steg, P.G.; Wijns, W. Stent thrombosis late after implantation of first-generation drug-eluting stents: A cause for concern. Circulation 2007, 115, 1440–1455. [Google Scholar] [CrossRef] [PubMed]
  23. Lagerqvist, B.; James, S.K.; Stenestrand, U.; Lindback, J.; Nilsson, T.; Wallentin, L.; Group, S.S. Long-term outcomes with drug-eluting stents versus bare-metal stents in Sweden. N. Engl. J. Med. 2007, 356, 1009–1019. [Google Scholar] [CrossRef] [PubMed]
  24. Nordmann, A.J.; Briel, M.; Bucher, H.C. Mortality in randomized controlled trials comparing drug-eluting vs. bare metal stents in coronary artery disease: A meta-analysis. Eur. Heart J. 2006, 27, 2784–2814. [Google Scholar] [CrossRef] [PubMed]
  25. Slottow, T.L.P.; Steinberg, D.H.; Roy, P.K.; Buch, A.N.; Okabe, T.; Xue, Z.; Kaneshige, K.; Torguson, R.; Lindsay, J.; Pichard, A.D.; et al. Observations and outcomes of definite and probable drug-eluting stent thrombosis seen at a single hospital in a four-year period. Am. J. Cardiol. 2008, 102, 298–303. [Google Scholar] [CrossRef] [PubMed]
  26. Cronin, C.J.; Ballenger, R.G. Alcohol use and negative consequences among American college students in West Germany. Int. J. Addict. 1991, 26, 1123–1136. [Google Scholar] [CrossRef] [PubMed]
  27. Joner, M.; Nakazawa, G.; Finn, A.V.; Quee, S.C.; Coleman, L.; Acampado, E.; Wilson, P.S.; Skorija, K.; Cheng, Q.; Xu, X.; et al. Endothelial cell recovery between comparator polymer-based drug-eluting stents. J. Am. Coll. Cardiol. 2008, 52, 333–342. [Google Scholar] [CrossRef] [PubMed]
  28. Fajadet, J.; Wijns, W.; Laarman, G.J.; Kuck, K.H.; Ormiston, J.; Munzel, T.; Popma, J.J.; Fitzgerald, P.J.; Bonan, R.; Kuntz, R.E.; et al. Randomized, double-blind, multicenter study of the Endeavor zotarolimus-eluting phosphorylcholine-encapsulated stent for treatment of native coronary artery lesions: Clinical and angiographic results of the ENDEAVOR II trial. Circulation 2006, 114, 798–806. [Google Scholar] [CrossRef] [PubMed]
  29. Bangalore, S.; Kumar, S.; Fusaro, M.; Amoroso, N.; Attubato, M.J.; Feit, F.; Bhatt, D.L.; Slater, J. Short- and long-term outcomes with drug-eluting and bare-metal coronary stents: A mixed-treatment comparison analysis of 117 762 patient-years of follow-up from randomized trials. Circulation 2012, 125, 2873–2891. [Google Scholar] [CrossRef] [PubMed]
  30. Kandzari, D.E.; Mauri, L.; Popma, J.J.; Turco, M.A.; Gurbel, P.A.; Fitzgerald, P.J.; Leon, M.B. Late-term clinical outcomes with zotarolimus- and sirolimus-eluting stents. 5-year follow-up of the ENDEAVOR III (a randomized controlled trial of the medtronic endeavor drug [ABT-578] eluting coronary stent system versus the cypher sirolimus-eluting coronary stent system in de novo native coronary artery lesions). JACC Cardiovasc. Interv. 2011, 4, 543–550. [Google Scholar] [PubMed]
  31. Camenzind, E.; Wijns, W.; Mauri, L.; Kurowski, V.; Parikh, K.; Gao, R.; Bode, C.; Greenwood, J.P.; Boersma, E.; Vranckx, P.; et al. Stent thrombosis and major clinical events at 3 years after zotarolimus-eluting or sirolimus-eluting coronary stent implantation: A randomised, multicentre, open-label, controlled trial. Lancet 2012, 380, 1396–1405. [Google Scholar] [CrossRef]
  32. Kirtane, A.J.; Leon, M.B.; Ball, M.W.; Bajwa, H.S.; Sketch, M.H., Jr.; Coleman, P.S.; Stoler, R.C.; Papadakos, S.; Cutlip, D.E.; Mauri, L.; et al. The “final” 5-year follow-up from the ENDEAVOR IV trial comparing a zotarolimus-eluting stent with a paclitaxel-eluting stent. JACC Cardiovasc. Interv. 2013, 6, 325–333. [Google Scholar] [CrossRef] [PubMed]
  33. Park, D.W.; Kim, Y.H.; Yun, S.C.; Kang, S.J.; Lee, S.W.; Lee, C.W.; Park, S.W.; Seong, I.W.; Lee, J.H.; Tahk, S.J.; et al. Comparison of zotarolimus-eluting stents with sirolimus- and paclitaxel-eluting stents for coronary revascularization: The ZEST (comparison of the efficacy and safety of zotarolimus-eluting stent with sirolimus-eluting and paclitaxel-eluting stent for coronary lesions) randomized trial. J. Am. Coll. Cardiol. 2010, 56, 1187–1195. [Google Scholar] [PubMed]
  34. Sabate, M.; Cequier, A.; Iniguez, A.; Serra, A.; Hernandez-Antolin, R.; Mainar, V.; Valgimigli, M.; Tespili, M.; den Heijer, P.; Bethencourt, A.; et al. Everolimus-eluting stent versus bare-metal stent in ST-segment elevation myocardial infarction (EXAMINATION): 1 year results of a randomised controlled trial. Lancet 2012, 380, 1482–1490. [Google Scholar] [CrossRef]
  35. Kaiser, C.; Galatius, S.; Erne, P.; Eberli, F.; Alber, H.; Rickli, H.; Pedrazzini, G.; Hornig, B.; Bertel, O.; Bonetti, P.; et al. Drug-eluting versus bare-metal stents in large coronary arteries. N. Engl. J. Med. 2010, 363, 2310–2319. [Google Scholar] [CrossRef] [PubMed]
  36. Kedhi, E.; Joesoef, K.S.; McFadden, E.; Wassing, J.; van Mieghem, C.; Goedhart, D.; Smits, P.C. Second-generation everolimus-eluting and paclitaxel-eluting stents in real-life practice (COMPARE): A randomised trial. Lancet 2010, 375, 201–209. [Google Scholar] [CrossRef]
  37. Stone, G.W.; Rizvi, A.; Newman, W.; Mastali, K.; Wang, J.C.; Caputo, R.; Doostzadeh, J.; Cao, S.; Simonton, C.A.; Sudhir, K.; et al. Everolimus-eluting versus paclitaxel-eluting stents in coronary artery disease. N. Engl. J. Med. 2010, 362, 1663–1674. [Google Scholar] [CrossRef] [PubMed]
  38. Jensen, L.O.; Thayssen, P.; Christiansen, E.H.; Maeng, M.; Ravkilde, J.; Hansen, K.N.; Hansen, H.S.; Krusell, L.; Kaltoft, A.; Tilsted, H.H.; et al. Safety and efficacy of everolimus-versus sirolimus-eluting stents: 5-year results from SORT OUT IV. J. Am. Coll. Cardiol. 2016, 67, 751–762. [Google Scholar] [CrossRef] [PubMed]
  39. Shiomi, H.; Kozuma, K.; Morimoto, T.; Igarashi, K.; Kadota, K.; Tanabe, K.; Morino, Y.; Akasaka, T.; Abe, M.; Suwa, S.; et al. Long-term clinical outcomes after everolimus- and sirolimus-eluting coronary stent implantation: Final 3-year follow-up of the randomized evaluation of sirolimus-eluting versus everolimus-eluting stent trial. Circ. Cardiovasc. Interv. 2014, 7, 343–354. [Google Scholar] [CrossRef] [PubMed]
  40. Serruys, P.W.; Silber, S.; Garg, S.; van Geuns, R.J.; Richardt, G.; Buszman, P.E.; Kelbaek, H.; van Boven, A.J.; Hofma, S.H.; Linke, A.; et al. Comparison of zotarolimus-eluting and everolimus-eluting coronary stents. N. Engl. J. Med. 2010, 363, 136–146. [Google Scholar] [CrossRef] [PubMed]
  41. Von Birgelen, C.; Basalus, M.W.; Tandjung, K.; van Houwelingen, K.G.; Stoel, M.G.; Louwerenburg, J.H.; Linssen, G.C.; Said, S.A.; Kleijne, M.A.; Sen, H.; et al. A randomized controlled trial in second-generation zotarolimus-eluting resolute stents versus everolimus-eluting Xience V stents in real-world patients: The TWENTE trial. J. Am. Coll. Cardiol. 2012, 59, 1350–1361. [Google Scholar] [PubMed]
  42. Serruys, P.W.; Garg, S.; Abizaid, A.; Ormiston, J.; Windecker, S.; Verheye, S.; Dubois, C.; Stewart, J.; Hauptmann, K.E.; Schofer, J.; et al. A randomised comparison of novolimus-eluting and zotarolimus-eluting coronary stents: 9-month follow-up results of the EXCELLA II study. EuroIntervention 2010, 6, 195–205. [Google Scholar] [PubMed]
  43. Tada, N.; Virmani, R.; Grant, G.; Bartlett, L.; Black, A.; Clavijo, C.; Christians, U.; Betts, R.; Savage, D.; Su, S.H.; et al. Polymer-free biolimus a9-coated stent demonstrates more sustained intimal inhibition, improved healing, and reduced inflammation compared with a polymer-coated sirolimus-eluting cypher stent in a porcine model. Circ. Cardiovasc. Interv. 2010, 3, 174–183. [Google Scholar] [PubMed]
  44. Urban, P.; Meredith, I.T.; Abizaid, A.; Pocock, S.J.; Carrie, D.; Naber, C.; Lipiecki, J.; Richardt, G.; Iniguez, A.; Brunel, P.; et al. Polymer-free drug-coated coronary stents in patients at high bleeding risk. N. Engl. J. Med. 2015, 373, 2038–2047. [Google Scholar] [CrossRef] [PubMed]
  45. Lansky, A.J.; Costa, R.A.; Mintz, G.S.; Tsuchiya, Y.; Midei, M.; Cox, D.A.; O′Shaughnessy, C.; Applegate, R.A.; Cannon, L.A.; Mooney, M.; et al. Non-polymer-based paclitaxel-coated coronary stents for the treatment of patients with de novo coronary lesions: Angiographic follow-up of the DELIVER clinical trial. Circulation 2004, 109, 1948–1954. [Google Scholar] [CrossRef] [PubMed]
  46. Moore, P.; Barlis, P.; Spiro, J.; Ghimire, G.; Roughton, M.; di Mario, C.; Wallis, W.; Ilsley, C.; Mitchell, A.; Mason, M.; et al. A randomized optical coherence tomography study of coronary stent strut coverage and luminal protrusion with rapamycin-eluting stents. JACC Cardiovasc. Interv. 2009, 2, 437–444. [Google Scholar] [CrossRef] [PubMed]
  47. King, L.; Byrne, R.A.; Mehilli, J.; Schomig, A.; Kastrati, A.; Pache, J. Five-year clinical outcomes of a polymer-free sirolimus-eluting stent versus a permanent polymer paclitaxel-eluting stent: Final results of the intracoronary stenting and angiographic restenosis—Test equivalence between two drug-eluting stents (ISAR-TEST) trial. Catheter. Cardiovasc. Interv. 2013, 81, E23–E28. [Google Scholar] [PubMed]
  48. Massberg, S.; Byrne, R.A.; Kastrati, A.; Schulz, S.; Pache, J.; Hausleiter, J.; Ibrahim, T.; Fusaro, M.; Ott, I.; Schomig, A.; et al. Polymer-free sirolimus- and probucol-eluting versus new generation zotarolimus-eluting stents in coronary artery disease: The intracoronary stenting and angiographic results: Test efficacy of sirolimus- and probucol-eluting versus zotarolimus-eluting stents (ISAR-TEST 5) trial. Circulation 2011, 124, 624–632. [Google Scholar] [PubMed]
  49. Carrie, D.; Berland, J.; Verheye, S.; Hauptmann, K.E.; Vrolix, M.; Violini, R.; Dibie, A.; Berti, S.; Maupas, E.; Antoniucci, D.; et al. A multicenter randomized trial comparing amphilimus—With paclitaxel-eluting stents in de novo native coronary artery lesions. J. Am. Coll. Cardiol. 2012, 59, 1371–1376. [Google Scholar] [PubMed]
  50. Joner, M.; Finn, A.V.; Farb, A.; Mont, E.K.; Kolodgie, F.D.; Ladich, E.; Kutys, R.; Skorija, K.; Gold, H.K.; Virmani, R. Pathology of drug-eluting stents in humans: Delayed healing and late thrombotic risk. J. Am. Coll. Cardiol. 2006, 48, 193–202. [Google Scholar] [CrossRef] [PubMed]
  51. Virmani, R.; Guagliumi, G.; Farb, A.; Musumeci, G.; Grieco, N.; Motta, T.; Mihalcsik, L.; Tespili, M.; Valsecchi, O.; Kolodgie, F.D. Localized hypersensitivity and late coronary thrombosis secondary to a sirolimus-eluting stent: Should we be cautious? Circulation 2004, 109, 701–705. [Google Scholar] [PubMed]
  52. Busch, R.; Strohbach, A.; Peterson, S.; Sternberg, K.; Felix, S. Parameters of endothelial function are dependent on polymeric surface material. Biomed. Tech. Berl. 2013. [Google Scholar] [CrossRef] [PubMed]
  53. Busch, R.; Strohbach, A.; Rethfeldt, S.; Walz, S.; Busch, M.; Petersen, S.; Felix, S.; Sternberg, K. New stent surface materials: The impact of polymer-dependent interactions of human endothelial cells, smooth muscle cells, and platelets. Acta Biomater. 2014, 10, 688–700. [Google Scholar] [CrossRef] [PubMed]
  54. Raber, L.; Kelbaek, H.; Ostoijc, M.; Baumbach, A.; Tuller, D.; von Birgelen, C.; Roffi, M.; Pedrazzini, G.; Kornowski, R.; Weber, K.; et al. Comparison of biolimus eluted from an erodible stent coating with bare metal stents in acute ST-elevation myocardial infarction (COMFORTABLE AMI trial): Rationale and design. EuroIntervention 2012, 7, 1435–1443. [Google Scholar] [CrossRef] [PubMed]
  55. Serruys, P.W.; Farooq, V.; Kalesan, B.; de Vries, T.; Buszman, P.; Linke, A.; Ischinger, T.; Klauss, V.; Eberli, F.; Wijns, W.; et al. Improved safety and reduction in stent thrombosis associated with biodegradable polymer-based biolimus-eluting stents versus durable polymer-based sirolimus-eluting stents in patients with coronary artery disease: Final 5-year report of the LEADERS (limus eluted from a durable versus erodable stent coating) randomized, noninferiority trial. JACC Cardiovasc. Interv. 2013, 6, 777–789. [Google Scholar] [PubMed]
  56. Stefanini, G.G.; Byrne, R.A.; Serruys, P.W.; de Waha, A.; Meier, B.; Massberg, S.; Juni, P.; Schomig, A.; Windecker, S.; Kastrati, A. Biodegradable polymer drug-eluting stents reduce the risk of stent thrombosis at 4 years in patients undergoing percutaneous coronary intervention: A pooled analysis of individual patient data from the ISAR-TEST 3, ISAR-TEST 4, and LEADERS randomized trials. Eur. Heart J. 2012, 33, 1214–1222. [Google Scholar] [PubMed]
  57. Byrne, R.A.; Kastrati, A.; Kufner, S.; Massberg, S.; Birkmeier, K.A.; Laugwitz, K.L.; Schulz, S.; Pache, J.; Fusaro, M.; Seyfarth, M.; et al. Randomized, non-inferiority trial of three limus agent-eluting stents with different polymer coatings: The intracoronary stenting and angiographic results: Test efficacy of 3 limus-eluting stents (ISAR-TEST-4) trial. Eur. Heart J. 2009, 30, 2441–2449. [Google Scholar] [CrossRef] [PubMed]
  58. Meredith, I.T.; Verheye, S.; Dubois, C.L.; Dens, J.; Fajadet, J.; Carrie, D.; Walsh, S.; Oldroyd, K.G.; Varenne, O.; El-Jack, S.; et al. Primary endpoint results of the EVOLVE trial: A randomized evaluation of a novel bioabsorbable polymer-coated, everolimus-eluting coronary stent. J. Am. Coll. Cardiol. 2012, 59, 1362–1370. [Google Scholar] [CrossRef] [PubMed]
  59. Smits, P.C.; Hofma, S.; Togni, M.; Vazquez, N.; Valdes, M.; Voudris, V.; Slagboom, T.; Goy, J.J.; Vuillomenet, A.; Serra, A.; et al. Abluminal biodegradable polymer biolimus-eluting stent versus durable polymer everolimus-eluting stent (COMPARE II): A randomised, controlled, non-inferiority trial. Lancet 2013, 381, 651–660. [Google Scholar] [CrossRef]
  60. Vlachojannis, G.J.; Smits, P.C.; Hofma, S.H.; Togni, M.; Vazquez, N.; Valdes, M.; Voudris, V.; Puricel, S.; Slagboom, T.; Goy, J.J.; et al. Long-term clinical outcomes of biodegradable polymer biolimus-eluting stents versus durable polymer everolimus-eluting stents in patients with coronary artery disease: Three-year follow-up of the COMPARE II (Abluminal biodegradable polymer biolimus-eluting stent versus durable polymer everolimus-eluting stent) trial. EuroIntervention 2015, 11, 272–279. [Google Scholar] [PubMed]
  61. Natsuaki, M.; Kozuma, K.; Morimoto, T.; Kadota, K.; Muramatsu, T.; Nakagawa, Y.; Akasaka, T.; Igarashi, K.; Tanabe, K.; Morino, Y.; et al. Biodegradable polymer biolimus-eluting stent versus durable polymer everolimus-eluting stent: A randomized, controlled, noninferiority trial. J. Am. Coll. Cardiol. 2013, 62, 181–190. [Google Scholar] [CrossRef] [PubMed]
  62. Saito, S.; Valdes-Chavarri, M.; Richardt, G.; Moreno, R.; Romo, A.I.; Barbato, E.; Carrie, D.; Ando, K.; Merkely, B.; Kornowski, R.; et al. A randomized, prospective, intercontinental evaluation of a bioresorbable polymer sirolimus-eluting coronary stent system: The CENTURY II (clinical evaluation of new terumo drug-eluting coronary stent system in the treatment of patients with coronary artery disease) trial. Eur. Heart J. 2014, 35, 2021–2031. [Google Scholar] [PubMed]
  63. Pilgrim, T.; Heg, D.; Roffi, M.; Tuller, D.; Muller, O.; Vuilliomenet, A.; Cook, S.; Weilenmann, D.; Kaiser, C.; Jamshidi, P.; et al. Ultrathin strut biodegradable polymer sirolimus-eluting stent versus durable polymer everolimus-eluting stent for percutaneous coronary revascularisation (BIOSCIENCE): A randomised, single-blind, non-inferiority trial. Lancet 2014, 384, 2111–2122. [Google Scholar] [CrossRef]
  64. Christiansen, E.H.; Jensen, L.O.; Thayssen, P.; Tilsted, H.H.; Krusell, L.R.; Hansen, K.N.; Kaltoft, A.; Maeng, M.; Kristensen, S.D.; Botker, H.E.; et al. Biolimus-eluting biodegradable polymer-coated stent versus durable polymer-coated sirolimus-eluting stent in unselected patients receiving percutaneous coronary intervention (SORT OUT V): A randomised non-inferiority trial. Lancet 2013, 381, 661–669. [Google Scholar] [CrossRef]
  65. Raungaard, B.; Jensen, L.O.; Tilsted, H.H.; Christiansen, E.H.; Maeng, M.; Terkelsen, C.J.; Krusell, L.R.; Kaltoft, A.; Kristensen, S.D.; Botker, H.E.; et al. Zotarolimus-eluting durable-polymer-coated stent versus a biolimus-eluting biodegradable-polymer-coated stent in unselected patients undergoing percutaneous coronary intervention (SORT OUT VI): A randomised non-inferiority trial. Lancet 2015, 385, 1527–1535. [Google Scholar] [CrossRef]
  66. Krucoff, M.W.; Kereiakes, D.J.; Petersen, J.L.; Mehran, R.; Hasselblad, V.; Lansky, A.J.; Fitzgerald, P.J.; Garg, J.; Turco, M.A.; Simonton, C.A.; et al. A novel bioresorbable polymer paclitaxel-eluting stent for the treatment of single and multivessel coronary disease: Primary results of the COSTAR (cobalt chromium stent with antiproliferative for restenosis) II study. J. Am. Coll. Cardiol. 2008, 51, 1543–1552. [Google Scholar] [CrossRef] [PubMed]
  67. Pandya, B.; Gaddam, S.; Raza, M.; Asti, D.; Nalluri, N.; Vazzana, T.; Kandov, R.; Lafferty, J. Biodegradable polymer stents vs. second generation drug eluting stents: A meta-analysis and systematic review of randomized controlled trials. World J. Cardiol. 2016, 8, 240–246. [Google Scholar] [CrossRef] [PubMed]
  68. Dudek, D.; Onuma, Y.; Ormiston, J.A.; Thuesen, L.; Miquel-Hebert, K.; Serruys, P.W. Four-year clinical follow-up of the ABSORB everolimus-eluting bioresorbable vascular scaffold in patients with de novo coronary artery disease: The ABSORB trial. EuroIntervention 2012, 7, 1060–1061. [Google Scholar] [CrossRef] [PubMed]
  69. Bowen, P.K.; Shearier, E.R.; Zhao, S.; Guillory, R.J.; Zhao, F.; Goldman, J.; Drelich, J.W. Biodegradable metals for cardiovascular stents: From clinical concerns to recent Zn-alloys. Adv. Healthc. Mater. 2016, 5, 1121–1140. [Google Scholar] [CrossRef] [PubMed]
  70. Haude, M.; Erbel, R.; Erne, P.; Verheye, S.; Degen, H.; Bose, D.; Vermeersch, P.; Wijnbergen, I.; Weissman, N.; Prati, F.; et al. Safety and performance of the drug-eluting absorbable metal scaffold (DREAMS) in patients with de-novo coronary lesions: 12 month results of the prospective, multicentre, first-in-man BIOSOLVE-I trial. Lancet 2013, 381, 836–844. [Google Scholar] [CrossRef]
  71. Erbel, R.; di Mario, C.; Bartunek, J.; Bonnier, J.; de Bruyne, B.; Eberli, F.R.; Erne, P.; Haude, M.; Heublein, B.; Horrigan, M.; et al. Temporary scaffolding of coronary arteries with bioabsorbable magnesium stents: A prospective, non-randomised multicentre trial. Lancet 2007, 369, 1869–1875. [Google Scholar] [CrossRef]
  72. Haude, M.; Ince, H.; Abizaid, A.; Toelg, R.; Lemos, P.A.; von Birgelen, C.; Christiansen, E.H.; Wijns, W.; Neumann, F.J.; Kaiser, C.; et al. Safety and performance of the second-generation drug-eluting absorbable metal scaffold in patients with de-novo coronary artery lesions (BIOSOLVE-II): 6 month results of a prospective, multicentre, non-randomised, first-in-man trial. Lancet 2016, 387, 31–39. [Google Scholar] [CrossRef]
  73. Tanimoto, S.; Bruining, N.; van Domburg, R.T.; Rotger, D.; Radeva, P.; Ligthart, J.M.; Serruys, P.W. Late stent recoil of the bioabsorbable everolimus-eluting coronary stent and its relationship with plaque morphology. J. Am. Coll. Cardiol. 2008, 52, 1616–1620. [Google Scholar] [CrossRef] [PubMed]
  74. Verheye, S.; Ormiston, J.A.; Stewart, J.; Webster, M.; Sanidas, E.; Costa, R.; Costa, J.R., Jr.; Chamie, D.; Abizaid, A.S.; Pinto, I.; et al. A next-generation bioresorbable coronary scaffold system: From bench to first clinical evaluation: 6- and 12-month clinical and multimodality imaging results. JACC Cardiovasc. Interv. 2014, 7, 89–99. [Google Scholar] [CrossRef] [PubMed]
  75. Serruys, P.W.; Ormiston, J.A.; Onuma, Y.; Regar, E.; Gonzalo, N.; Garcia-Garcia, H.M.; Nieman, K.; Bruining, N.; Dorange, C.; Miquel-Hebert, K.; et al. A bioabsorbable everolimus-eluting coronary stent system (ABSORB): 2-year outcomes and results from multiple imaging methods. Lancet 2009, 373, 897–910. [Google Scholar] [CrossRef]
  76. Ellis, S.G.; Kereiakes, D.J.; Metzger, D.C.; Caputo, R.P.; Rizik, D.G.; Teirstein, P.S.; Litt, M.R.; Kini, A.; Kabour, A.; Marx, S.O.; et al. Everolimus-eluting bioresorbable scaffolds for coronary artery disease. N. Engl. J. Med. 2015, 373, 1905–1915. [Google Scholar] [CrossRef] [PubMed]
  77. Serruys, P.W.; Chevalier, B.; Dudek, D.; Cequier, A.; Carrie, D.; Iniguez, A.; Dominici, M.; van der Schaaf, R.J.; Haude, M.; Wasungu, L.; et al. A bioresorbable everolimus-eluting scaffold versus a metallic everolimus-eluting stent for ischaemic heart disease caused by de-novo native coronary artery lesions (ABSORB II): An interim 1-year analysis of clinical and procedural secondary outcomes from a randomised controlled trial. Lancet 2015, 385, 43–54. [Google Scholar] [PubMed]
  78. Zhang, X.L.; Zhu, L.; Wei, Z.H.; Zhu, Q.Q.; Qiao, J.Z.; Dai, Q.; Huang, W.; Li, X.H.; Xie, J.; Kang, L.N.; et al. Comparative efficacy and safety of everolimus-eluting bioresorbable scaffold versus everolimus-eluting metallic stents: A systematic review and meta-analysis. Ann. Intern. Med. 2016, 164, 752–763. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary of partially bioresorbable coronary stents.
Table 1. Summary of partially bioresorbable coronary stents.
StentScaffold MaterialDrugPolymer CoatingStrut Thickness (μm)Coating Thickness (μm)ManufacturerClinical Study
AxxessNitinolBiolimus A9PDLLA15215BiosensorsDIVERGE
BioMimeCo-CrSirolimusPLLA/PLGA652Meril Life SciencesmeriT-1
BioMatrixSSBiolimus A9PDLLA11210BiosensorsLEADERS; SORTOUT VI
ComboSSSirolimusPDLLA/PLGA1003–5OrbusNeichREMEDEE
DESyne BDCo-CrNovolimusPLLA81<3Elixir MedicalEXCELLA BD
InfinniumSSPaclitaxelPLLA, PLGA, PCL, PVP804–5Sahajanand MecialSIMPLE II
MiStentCo-CrCrystalline SirolimusPLGA645/15MicellDESSOLVE I
NoboriSSBiolimus A9PDLLA11210TerumoNEXT; SORTOUT V; COMPARE-II
OrsiroCo-CrSirolimusPLLA603.5/7.5BiotronikBIOSCIENCE
SupralimusCo-CrSirolimusPLLA, PLGA, PCL, PVP804–5Sahajanand MecialSERIES I
SynergyPt-CrEverolimusPLGA744Boston ScientificEVOLVE
UltimasterCo-CrSirolimusPDLLA/PCL8015TerumoCENTURY II
Yukon Choice PCSSSirolimusPDLLA875TransluminaISAR-TEST-4
Note: Co-Cr = Cobalt-Chrome; PCL = poly(l-lactide co-e-caprolactone); PDLLA = poly(d,l)-lactic acid; PLGA = poly(lactide-co-glycolide); PLLA = poly-l-lactic acid; Pt-Cr = Platinum-chrome; PVP = polyvinylpyrrolidone; SS = Stainless Steel.
Table 2. Summary of fully bioresobable coronary stents with Conformite′ Europe′ enne.
Table 2. Summary of fully bioresobable coronary stents with Conformite′ Europe′ enne.
StentScaffold MaterialDrugPolymer CoatingStrut Thickness (μm)ManufacturerClinical Study
DREAMSMgPaclitaxelPLGA125BiotronikBIOSOLVE-1
DREAMS 2GMgSirolimusPLLA150Biotronik
Absorb BVSPLLAEverolimusPDLLA156Abbot VascularABSORB
DESolvePLLAMyolimusPLLA150Elixir MedicalDESOLVE
DESolve 100PLLANovolimusPLLA100Elixir Medical
Note: Mg = Magnesium; PDLLA = poly(d,l)-lactic acid; PLGA = poly(lactide-co-glycolide); PLLA = poly-l-lactic acid.

Share and Cite

MDPI and ACS Style

Ho, M.-Y.; Chen, C.-C.; Wang, C.-Y.; Chang, S.-H.; Hsieh, M.-J.; Lee, C.-H.; Wu, V.C.-C.; Hsieh, I.-C. The Development of Coronary Artery Stents: From Bare-Metal to Bio-Resorbable Types. Metals 2016, 6, 168. https://doi.org/10.3390/met6070168

AMA Style

Ho M-Y, Chen C-C, Wang C-Y, Chang S-H, Hsieh M-J, Lee C-H, Wu VC-C, Hsieh I-C. The Development of Coronary Artery Stents: From Bare-Metal to Bio-Resorbable Types. Metals. 2016; 6(7):168. https://doi.org/10.3390/met6070168

Chicago/Turabian Style

Ho, Ming-Yun, Chun-Chi Chen, Chao-Yung Wang, Shang-Hung Chang, Ming-Jer Hsieh, Cheng-Hung Lee, Victor Chien-Chia Wu, and I-Chang Hsieh. 2016. "The Development of Coronary Artery Stents: From Bare-Metal to Bio-Resorbable Types" Metals 6, no. 7: 168. https://doi.org/10.3390/met6070168

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop