Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Mar 21.
Published in final edited form as: Circ Cardiovasc Interv. 2012 Apr;5(2):296–304. doi: 10.1161/CIRCINTERVENTIONS.111.965426

Cardiovascular Devices and Platelet Interactions Understanding the Role of Injury, Flow, and Cellular Responses

Jesse W Rowley 1, Aloke V Finn 1, Patricia A French 1, Lisa K Jennings 1, Danny Bluestein 1, Peter L Gross 1, Jane E Freedman 1, Steven R Steinhubl 1, Guy A Zimmerman 1, Richard C Becker 1, Harold L Dauerman 1, Susan S Smyth 1; The 2011 Platelet Colloquium Participants1,*
PMCID: PMC3605036  NIHMSID: NIHMS434438  PMID: 22511738

The objectives of the 2011 Platelet Colloquium were to gain a better understanding of the role of platelets and inflammatory cells in the response to cardiovascular device–associated injury; to describe the relationship between cardiac devices, shear stress, and alterations in platelet biology and function; and to review evidence derived from in vitro, in vivo, and clinical investigations supporting therapies to modify platelet and inflammatory cell–associated responses to device-related injury.

Accordingly, this review summarizes the evidence for translating basic scientific concepts underlying circulatory and device-associated vascular injury to the clinical development and use of cardiac devices, supplemented by findings from the literature. Insights for platelet receptor activation, inflammatory cell biology, endothelial dysregulation, and the effects of biomechanical stress on coagulation and hemostatic proteins were examined to better determine the pathobiology, incidence, and possible prevention of adverse events in patients receiving cardiac devices.

Shear Stress and Platelet Activation

Hemodynamics play a key role in thrombus formation. Pathophysiological shear stress induces activation of platelets and the endothelium, increases platelet–leukocyte interactions, and promotes thrombin generation. The initial tethering of rolling activated platelets, subsequent stable adhesion, and aggregation each are influenced by shear stress on the vessel wall, which can range from 11.4 to 30.4 dyne/cm2 in large arteries and up to 380 dyne/cm2 at critical areas of arterial stenosis.1 Along a diseased vessel, shear forces will vary dramatically within short distances, increasing as the lumen narrows and then decreasing in the poststenotic vascular segment.2

Several platelet membrane proteins serve as mechanosensors, most notably glycoprotein (GP) Ib and its ligand, von Willebrand factor (vWF).3 vWF circulates as highly adhesive multimers (ultralarge vWF) that are cleaved by a disintegrin and metalloproteinase with thrombospondin type 1 motif member 13 (ADAMTS-13). Depletion or accumulation of ultralarge vWF, as a consequence of inappropriate ADAMTS-13 proteolysis, results in pathological bleeding (as in some patients with type 2A von Willebrand disease) or microvascular thrombosis (as with thrombotic thrombocytopenic purpura), respectively.

Shearing forces regulate vWF multimer size by unfolding a Tyr1605–Met1606 bond within the vWF-A2 domain such that it becomes a substrate for ADAMTS-13 cleavage.36 Platelet GP Ib responds to higher shear with increased vWF binding and intracellular signaling, in part by generating a calcium flux7 that together with phosphoinositide 3-kinase8 triggers integrin αIIbβ3 activation.7,9 These signals convert transient platelet surface interactions into stable integrin-mediated adhesion. During shear-induced platelet activation in vitro, released adenosine diphosphate (ADP) plays a fundamental role in this important process.

Temporal shear gradients promote cooperative signaling between the purinergic P2Y1 receptor and integrin αIIbβ3,10,11 which in turn contributes to the development of P2Y12-dependent stable platelet thrombi.11 Shear stress dissociates integrin αIIbβ3–cytoskeletal protein interactions, including associations with α-actin and myosin heavy chains,12 although the consequences of this disruption are unknown.

Several factors stabilize formed platelet aggregates. The intercellular space between aggregating platelets provides a protective environment for accumulation of ADP, thrombin, thromboxane A2, Gas-6, and exodomains of platelet surface proteins such as GP Ib, CD62, and soluble CD40 ligand.13 The contributions of the αIIβ1, P2Y1, P2Y12, and αIIbβ3 integrins are also critical to development of stable thrombi.1417 Accordingly, agents targeting these latter receptors can reduce existing thrombus burden,18,19 although increased intracoronary dose levels may be necessary to dissolve rather than prevent thrombus.20

The effects of shear on platelets might extend beyond acute activation and thrombosis. Platelets have played a role in atherosclerosis initiation and/or progression in animal models, although the exact role of platelet activation is still uncertain.21,22 Inhibition of platelet activation, vWF deficiency, GP Ib blockade, and αIIb deficiency all have shown protective effects against atherogenesis in mice.2325 In a study of recombinant platelet GP Ib as the targeting entity on lipid-shelled decafluorobutane microbubbles to detect “activated” vWF and other GP Ib ligands, Lindner et al26 detected microbubble accumulation along thrombi formed by ex vivo perfusion of human blood over a collagen surface and along atherosclerotic vessels. In atherosclerotic mice, signal strength likewise was greatest in areas with severe atherosclerotic lesions and in regions associated with platelet accumulation.

Oxidation of vWF by oxidants derived from inflammatory leukocytes can render vWF more stable, with enhanced platelet binding and aggregatory affects.27 The potential combined effects of shear and inflammation on vWF-platelet binding highlights the complex interplay among shear stress, inflammation, and platelet–leukocyte activation (Figure 1). Other shear/platelet/inflammatory interactions are undoubtedly at play at the site of atherosclerotic lesions, such as the formation of platelet–leukocyte heterotypic aggregates induced by shear-mediated P-selectin induction in platelets.28 Other effects of shear on vascular inflammation are further examined elsewhere.29

Figure 1.

Figure 1

Complex interplay among shear stress, inflammation, and platelet–leukocyte activation. vWF indicates von Willebrand factor; RBC, red blood cell; PSGL, P-selectin glycoprotein ligand; GP, glycoprotein; PKC, protein kinase C; TGF, transforming growth factor; and ADP, adenosine diphosphate.

In Vivo and In Vitro Modeling of Platelet Function Under Shear

Most of the classic methods for testing platelet function do not accurately reflect in vivo events occurring along vascular surfaces in the presence of flowing blood. Even putative markers for “in vivo platelet activation”—platelet microparticles, β-thromboglobulin, and thromboxane— exhibit poor reproducibility.30 Tests of platelet “activatability,” such as classic aggregation testing, flow cytometry, VerifyNow point-of-care testing, and the MultiPlate system, rely on low-shear settings that may not accurately mimic the high-shear environment present within diseased blood vessels. Several strategies to model and monitor platelets under more physiological conditions, including animal models, imaging in humans, and in vitro flow are now being used, a selection of which is summarized in Table 1.

Table 1.

Advantages and Disadvantages of Selected In Vitro and In Vivo Methods Used to Measure Platelet Activation*

Method Advantages Disadvantages
Confocal imaging in mice of in vivo platelet activation and thrombus formation Genetically manipulated, environmentally controlled, lower cost, can visualize multiple parameters at once in real time in physiological context Species differences in cell size, molecular signatures, vessel size, shear; not amenable to device testing
Large-animal studies Environmentally controlled, more similar cell sizes and shears (animal-dependent), can study devices/drugs in physiological context Species differences molecular signatures, higher cost, limited availability
Contrast ultrasound with microbubbles targeting platelet ligands and activation markers Potential translation to humans, can study devices/drugs in physiological context Higher cost, limited availability
Indices of platelets (from lesion) coupled with 3-dimensional coronary angiography In humans, can study devices/drugs in physiological context Higher cost, indirect measurement after blood draw, limited availability
In vitro chromogenic assays of thrombin formation coupled with hemodynamic shearing device Low cost, wide availability, no requirement for humans or animals, better than static assays, can perform real-time measurements, rapid Cannot study in physiological context
In vitro flow chamber studies Low cost, wide availability, no requirement for humans or animals, can test many variables from single sample, better than static assays, rapid Cannot study in physiological context
*

Methods discussed in the text; not meant to be comprehensive.

Newer technologies permit real-time measurement of in vivo thrombus development in animal models, by means of a wide-field fluorescence microscope fitted with a confocal head.31 Using these technologies, several hallmarks of platelet activation have been observed (Table 2). In one instance, pretreatment of mice with 1.25 μg/kg aspirin by mouth <1 hour before laser injury of mouse cremaster muscle arterioles did not affect platelet accumulation (thrombus volume) but reduced activation of integrin αIIbβ3 by 67% as assessed by high-speed, 2-color confocal microscopy.32 Such technologies may further our understanding of platelet activation in vivo and thus allow the design of more effective inhibitors.

Table 2.

Hallmarks of Platelet Activation Seen With New In Vivo Technologies

Hallmark Physiology Marker
αIIbβ3 Integrin increase and activation Stable aggregation Antibody to αIIb or activated ligand bound to integrin
Dense granule secretion ADP, serotonin release for amplification Lumiaggregometry or 14[C]5HT release
α-Granule secretion P-selectin for cell interaction Antibody to P-selectin
Procoagulants (V) Prothrombin activation
Anti/angiogenic factors PF4
Phosphatidylserine expression Procoagulant surface Annexin/Diannexin
Platelet–leukocyte interaction Inflammatory/thrombotic signaling Genetically labeled platelets/leukocytes and their antibodies
Receptor shedding Downregulation? Antibody

ADP indicates adenosine diphosphate.

Strategies for monitoring interactions between shear and antiplatelet drug effects have been developed for humans. For example, Yong et al33 obtained blood from the coronary arteries proximal and distal to coronary lesions and from the coronary sinus in patients with stable coronary artery disease taking both aspirin and clopidogrel. After estimating the shear stress from 3-dimensional coronary angiographic images of these arteries, they assessed the effects of stenosis severity and shear stress on in vivo activation of platelets and monocytes. They found increased platelet P-selectin expression, platelet–monocyte aggregation, and monocyte CD11b expression, but integrin αIIbβ3 activation and soluble P-selectin did not correlate with stenosis severity and shear stress. Shear-related intracoronary platelet and monocyte activation might represent a therapeutic target distinct from conventional antiplatelet agents.

Modeling Platelet-Device Interactions

Thrombotic complications with cardiovascular devices remain a critical limitation to their long-term use. Device-induced shear forces can enhance hemostatic response through chronic activation of platelets, with a known dose-time response in terms of accumulated stress.34 Because of the relative infrequency of thrombosis with current models of stents, however, demonstrating its reduction in clinical trials of newer stent platforms will probably require tens of thousands of patients.

Alternatively, in vitro modeling techniques for testing device-mediated thrombogenicity, coupled with virtual design modifications in an iterative approach, could be incorporated during device research and development. Such a method would optimize the thrombogenic performance of the device before prototypes are tested in costly preclinical and clinical trials.

Rather than testing device-mediated thrombogenicity in clinical trials or animal models, an attractive alternative strategy might be to harness in vitro modeling techniques that could be incorporated during device research and development. Such a method would use virtual design modifications in an iterative approach to optimize the thrombogenic performance of the device before prototypes are built and tested in costly preclinical and clinical trials.

A recent study used in vitro flow-chamber studies to compare platelet aggregation after exposure to drug-eluting stents (DES) and bare metal stents (BMS) with variable strut thickness. The thin-polymer/thin-strut DES elicited less platelet activation and aggregation than did the thick-strut DES; more important, the thin-polymer/thin-strut DES in this model provoked less platelet activation/aggregation than did the BMS platforms.35 Such in vitro modeling allows rational development and examination of small changes in device designs as a method for guiding future trial designs.

Another example of effective in vitro modeling uses a real-time chromogenic assay27 to measure thrombin formation, which can be applied to understand the impact of devices on the thrombogenicity of flowing blood. Thrombin formation is monitored as blood flows past mechanical devices, and the extent of thrombin generation relates directly to the platelet activation and aggregation induced.3638 Chromogenic monitoring of thrombin generation can be coupled with a novel hemodynamic shearing device (HSD) to expose platelets to dynamic shear-stress waveforms as extracted from advanced numeric simulations of blood flow through the device. This shearing device can allow comprehensive evaluation of the thrombogenic potential of devices as a potential surrogate for initial animal or human testing (Figure 2).39 This system can identify “hot spot” regions within the device during distinct flow phases and compute flow trajectories and stress load histories within these regions.

Figure 2.

Figure 2

Device thrombogenicity emulator. “Hot spot” trajectories from numeric simulations (shown, blood flow through a bileaflet mechanical heart valve, top) are translated into shear stress–loading waveforms τ(t). These waveforms are then programmed into the hemodynamic shearing device (bottom left), platelets are repeatedly exposed to them, and the resultant activity is measured using a platelet activity state assay (bottom right). Adapted from Bluestein et al,39 with permission. VWF indicates von Willebrand factor.

Designs can then be iteratively modified to reduce thrombogenic potential computed in the virtual domain and the resultant stress-loading waveforms programmed and tested in the HSD. This system has been termed the “device thrombogenicity emulator” (DTE). Although primarily used to study mechanical circulatory support devices and bileaflet mechanical heart valves,40,41 in vitro assessment of hemodynamic shearing forces and subsequent platelet activation might be an efficient means of making iterative changes in other potentially thrombogenic cardiac and arterial devices, including stents. The HSD can study the interaction between flowing platelets and cell-cultured endothelium.42 The system is operated under emulated shear stress loading conditions extracted from numeric simulations of blood flow in various device configurations. In this manner, the HSD could allow expeditious assessment of iterative device designs with respect to platelet activation and thrombogenicity.35

Role of Platelet Biology in Device-Induced Injury

In addition to mediating acute arterial thrombosis, platelets play a critical role in the response to and recovery from vascular injury. Activated platelets interact with leukocytes after device deployment43 and promote the recruitment of leukocytes to inflamed endothelium. Platelets contain variable levels of mRNAs, premRNAs, and microRNAs and a repertoire of translational pathways for posttranscriptional synthesis of new proteins.4446 Both genetic and environmental factors influence the expression patterns of transcripts and their corresponding protein products. For example, the differential expression of several inflammatory transcripts correlates with body mass index and cardiovascular disease.47 Analyzing acute and prolonged changes in the platelet transcriptome and proteome might prove fruitful in identifying causes for differences in outcomes of device-mediated injury.

Experimental models of restenosis have shown direct correlations between the frequency and severity of cyclic flow variations, platelet aggregation, and neointimal proliferation.4850 Moreover, in animal models, administration of agents that block the ADP, thromboxane A2, and serotonin pathways49 and agents that inhibit platelet-monocyte interactions50 results in significantly larger residual luminal areas. In humans, platelet reactivity measured before percutaneous coronary intervention (PCI) has been linked to repeat revascularization at 90 days51 and target-vessel revascularization at 7 months.52

Despite the solid mechanistic underpinning for antiplatelet agents in modulating the response to vascular injury after PCI, large clinical investigations have yielded mostly disappointing results in terms of clinical outcomes.5355 Even abciximab, which substantially reduced the incidence of target vessel revascularization in 2 pivotal trials,56,57 has failed to show significant benefit in reducing restenosis.5860 Clearly, inhibition of platelet aggregation is not the sole answer to improving outcomes after vascular trauma.

Several studies have investigated the ability of cilostazol, a phosphodiesterase III inhibitor approved in the United States for treatment of intermittent claudication, to limit neointimal proliferation after stenting. A recent meta-analysis of 10 randomized trials (n=2809 patients overall) reported a significant, 48% decrease in angiographic restenosis when cilostazol was added to aspirin and thienopyridine therapy, without major differences between BMS or DES cohorts.61 Target lesion revascularization was reduced by 62%, but subacute stent thrombosis and major bleeding did not differ significantly. An initial randomized trial of a new, cilostazoleluting stent reported significantly reduced in-stent late loss and restenosis versus a paclitaxel-eluting stent, with no stent thrombosis or myocardial infarction (MI) in any of the 111 patients at 8 months.62 This technology might represent a promising new type of DES system.

Alternatively, based on the mechanistic role of inflammation in vascular injury,63 several investigations have centered on the antiplatelet and anti-inflammatory effects of statins as a means to facilitate recovery from vascular injury.6466 Plasma levels of soluble CD40 ligand and prothrombin fragment F1.2 were significantly reduced among hypercholesterolemic patients who received atorvastatin for 3 days (n=15) versus diet (n=15), and atorvastatin was associated with a dose-dependent, significant decrease in platelet CD40 ligand expression in vitro among both the hypercholesterolemic patients and 20 healthy control subjects.64 Previous animal models have shown atheroma formation and regression to be CD40 ligand-dependent.67 These mechanistic data may partly explain the findings of the REVERSing atherosclerosis with Aggressive Lipid lowering (REVERSAL) trial and A Study To Evaluate the effect of Rosuvastatin On Intravascular ultrasounD (ASTEROID), in which patients receiving intensive statin therapy (atorvastatin 80 mg or rosuvastatin 40 mg daily, respectively) showed dose-dependent, significant decreases in atheroma volume on intravascular ultrasonography.65,66

Recent studies examining high-dose statin therapy given 24 hours before PCI suggest that short-term 3-hydroxy-3-methyl-glutayl coenzyme A (HMG-CoA) reductase inhibition might reduce ischemic events.68,69 The mechanisms responsible are unknown but are probably not mediated primarily by reduced low-density lipoprotein C levels. The early beneficial effects of statins on ischemic events might reflect antiplatelet and/or anti-inflammatory properties. Statin therapy has been associated with lower urinary thromboxane levels,70 a marker of activated circulating platelets, and reduced ex vivo shear-induced platelet activation.71 However, not all studies have found an effect of statins on platelet function.72

Histopathologic Insights Into Acute and Delayed Healing Responses After BMS/DES Implantation

Although platelets have played a critical role in responses to vascular injury in several animal models, available antiplatelet therapies generally have failed to show a favorable effect on restenosis in clinical trials. This does not mean that platelet-mediated growth factor and inflammatory responses do not play large roles in the processes of arterial injury and repair. Both balloon dilatation and stenting result in endothelial denudation, whereby adhesion molecules such as P-selectin mediate platelet–leukocyte– endothelial cell interactions. These interactions spur platelets to release cytokines and growth factors that mediate smooth muscle cell migration and proliferation.7375

The mechanisms for development of intimal hyperplasia can differ after angioplasty versus stenting. For example, unlike angioplasty, which initiates early neutrophil recruitment, stenting tends to result in prolonged macrophage recruitment.76 Nonetheless, the downstream pathways involved in arterial injury all initiate the cell cycle regulated by cycling-dependent kinases. Agents that inhibit these kinases, such as rapamycin, are extremely effective in inhibiting restenosis, especially when delivered locally. This phenomenon highlights the importance of common molecular signaling pathways that initiate the process of intimal formation.

Technical improvements in PCI and stents have reduced restenosis rates from ≈30% in the 1980s to <10% today. The introduction of BMS in the 1990s represented a significant advance, with additional refinement in the 2000s through the debut of DES. Even within the DES category, designs have continued to evolve in an effort to optimize outcomes. Stent strut widths, for example, have decreased from 97 to 140 μm with the first generation of devices (Cypher, Taxus) to 81 to 91 μm with the second (Endeavor, Xience). Thinner stent struts may be associated with improved clinical outcomes, but further study is needed for clarification.58,7779

The mechanisms of late and very late stent thrombosis (LST) are unknown. Clinical trials using prasugrel or ticagrelor clearly demonstrate a role of P2Y12 receptors in LST.80,81 In analyses of autopsy and clinical registry data, DES have been associated with delayed coronary arterial healing, as evidenced by persistent fibrin deposition and reduced endothelialization, and an increased risk of LST compared with BMS.82,83 In 1 case-control analysis of data from 62 lesions in the CVPath Institute's registry, endothelialization was the best independent predictor of thrombosis >30 days after stenting, which in turn was best correlated with the ratio of uncovered to total stent struts per section (odds ratio for thrombus in a stent with a >30% ratio of uncovered to total stent struts/section, 9.0; 95% confidence interval, 3.5–22).84

One potential mechanism for LST, at least among some first-generation stents, relates to hypersensitivity reactions85 that appeared to be limited to sirolimus-eluting devices.85 However, stent malapposition secondary to excessive fibrin deposition appeared to be an exclusive characteristic of paclitaxel-eluting stents in analysis of 230 lesions from the CVPath registry.86 Another analysis of 51 patients from the same registry implicated stenting for acute MI in development of LST.87 Compared with patients who had stable angina with thick-cap fibroatheromas, those with an MI culprit site had significantly smaller neointimal thickness (0.04 versus 0.11 mm) and greater proportions of uncovered struts (49% versus 9%), struts with fibrin deposition (63% versus 36%), and struts with inflammation (35% versus 17%; all P≤0.01).87 These findings emphasize the importance of plaque morphology to arterial responses after DES implantation.

Neoatherosclerotic changes developing within the neointima after stenting have also been proposed as a mechanism for LST. Atherosclerosis develops earlier and more often within the neointima among patients receiving DES versus BMS.88 In patients undergoing follow-up intravascular imaging at least 20 months after DES placement, features consistent with high-risk plaques, such as rupture and thrombi within the neointima, have been noted.88 This unstable neoatherosclerosis in DES-treated patients probably contributes to late thrombotic events.

The number of uncovered struts might serve as a surrogate measure of epithelialization/neointimal coverage after stenting and thus might inform the duration of antiplatelet therapy. Optical coherence tomography (OCT) has been proposed as a noninvasive method of assessing this surrogate. In a study of 243 patients undergoing DES implantation, neointimal hyper-plasia morphology on OCT correlated moderately with simultaneous intravascular ultrasound findings obtained a mean 12 months after stenting, but assessments differed in 31% of lesions.89 Another analysis of 622 OCT-histology-matched samples from 84 stents of 4 types showed good correlations between methods in neointimal area, luminal area, and neointimal thickness.90 Overall, the proportions of uncovered struts identified by OCT and histology were similar, but estimations varied significantly between methods at neointimal thicknesses of 20 to 80 μm. Whether OCT-determined strut coverage can actually predict clinical outcomes remains to be determined.

Modeling in large animals91 and humans92 has correlated shear stress with neointimal proliferation in stents. Human studies comparing BMS and DES identified shear stress as an independent predictor of neointimal thickness,93 with sirolimus-eluting stents (but not paclitaxel-eluting stents) reducing the effect of shear stress on neointimal proliferation.

Inflammatory and platelet activation reactions, including stent thrombosis, might be a lesser problem with newer DES.94 In a flow-loop model assessing in vitro thrombogenicity of DES, drug/polymer coatings did not enhance thrombogenicity compared with their BMS counterparts, although stent positioning and design were determinants of stent thrombogenicity.35 More recent randomized trials also have failed to show a clear increase in LST risk with DES versus BMS. For example, the Efficacy of Xience/Promus versus Cypher in rEducing Late Loss after stENTing (EXCELLENT) trial comparing Xience and BMS suggests that second-generation DES might carry an even lower 1-year rate of LST than do BMS.95 Of note, this trial was a noninferiority study based on late loss; it was unpowered to examine “hard” end points such as stent thrombosis or MI. Thus, definitive conclusions cannot be made about the safety of one device versus another at present. Biocompatible polymers, and disappearing polymers/stents, might minimize some of the concerns regarding pathological platelet activation and inflammation.95,96

Summary

Shear influences platelet adhesion and activation through mechanisms that include altering vWF binding and αIIbβ3/purinergic signaling interactions. Shear can potentially promote inflammation and atherogenesis through platelet-mediated processes. New methods to study platelet function under shear conditions and in more physiological contexts should be incorporated to guide the design of future devices and drug therapies. Some of these methods include in vivo imaging, in vitro flow models, and in vitro modeling/computer-assisted iterative device design.

Improving currently used antiplatelet agents is not the sole answer for reducing vascular injury after PCI. Cilostazol and statins are promising examples of agents that have effects beyond inhibition of platelet activation. In addition, LST, which has been associated with both stent design and pathological changes (such as endothelialization and neoathero-sclerosis within the neointima), is becoming a less critical issue with DES because of the use of DAPT and newer stent designs.

Conclusions

Our knowledge regarding interactions among platelets, vascular flow conditions, and cardiovascular device characteristics continues to evolve. Simultaneously, we are better appreciating the role of inflammatory cells in the response to cardiac device-associated injury. Novel in vitro and in vivo techniques now permit accurate assessment of the thrombogenicity of new device designs and might aid in development of therapeutic targets distinct from conventional antiplatelet agents. By focusing on the mechanistic aspects of cardiac devices and the vasculature, and on the inflammatory response, the goal of translating promising interventions to optimal patient care can be achieved more rapidly and efficiently.

Acknowledgments

Sources of Funding The 2011 Platelet Colloquium and development of this report were supported by unrestricted grants from Abbott Vascular, Inc, Redwood City, CA; AstraZeneca, PLC, Wilmington, DE; Boston Scientific Corporation, Natick, MA; Daiichi Sankyo, Inc, and Lilly USA LLC Partnership, Parsippany, NJ; Medtronic Cardiovascular, Minneapolis, MN; Regado Biosciences, Inc, Durham, NC; St Jude Medical, Inc, St Paul, MN; and The Medicines Company, Parsippany, NJ. This material is the result of work supported with the resources and use of the facilities at the Lexington Veterans Affairs Medical Center (Dr Smyth). The contents do not represent the views of the Department of Veterans Affairs or the US Government.

Footnotes

Disclosures Dr Finn has received grants from Boston Scientific, Medtronic, and St Jude Medical and has consulted for Abbott Vascular. Patricia French has consulted for Regado Biosciences. Dr Jennings has received grants from Atrium Medical, AstraZeneca, and Merck; has participated in speaker's bureaus for BMS/sanofi-aventis and Merck; has received honoraria from AstraZeneca and Merck; and has consulted for BMS/sanofi-aventis, Merck, and Portola Pharmaceuticals. Dr Gross has received research grants from the Canadian Institutes for Health Research, the Heart and Stroke Foundation of Ontario, and Canadian Blood Services; has participated in speaker's bureaus for Bayer and Leo Pharma; and has been an expert witness for Apotex. Dr Steinhubl is a former employee of The Medicines Company. Dr Zimmerman has received grants from Boehringer-Ingelheim and NovoNordisk. Dr Becker has received grants from AstraZeneca, Bayer, and Regado Biosciences; has received honoraria from Johnson & Johnson and Regado Biosciences; and has consulted for Daiichi/Lilly and Boehringer-Ingelheim. Dr Dauerman has received research grants from Abbott Vascular, Medtronic, and MDS Scientific; has consulted for Abbott Vascular, Medtronic, MDS Scientific, Novartis, The Medicines Company, Gilead, and St Jude Medical; and has served as an expert defense witness in New Hampshire. Dr Smyth has received grants from AstraZeneca, The Medicines Company, and Boehringer-Ingelheim and has consulted for BMS/sanofi-aventis.

The online-only Data Supplement is available with this article at http://circinterventions.ahajournals.org/lookup/suppl/doi:10.1161/CIRCINTERVENTIONS.111.965426/-/DC1.

References

  • 1.Slack SM, Turitto VT. Fluid dynamic and hemorheologic considerations. Cardiovasc Pathol. 1993;2:11S–21S. [Google Scholar]
  • 2.Strony J, Beaudoin A, Brands D, Adelman B. Analysis of shear stress and hemodynamic factors in a model of coronary artery stenosis and thrombosis. Am J Physiol. 1993;265:H1787–H1796. doi: 10.1152/ajpheart.1993.265.5.H1787. [DOI] [PubMed] [Google Scholar]
  • 3.Jakobi AJ, Mashaghi A, Tans SJ, Huizinga EG. Calcium modulates force sensing by the von Willebrand factor A2 domain. Nat Commun. 2011;2:385. doi: 10.1038/ncomms1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Gao W, Anderson PJ, Sadler JE. Extensive contacts between ADAMTS13 exosites and von Willebrand factor domain A2 contribute to substrate specificity. Blood. 2008;112:1713–1719. doi: 10.1182/blood-2008-04-148759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Zhang Q, Zhou Y-F, Zhang C-Z, Zhang X, Lu C, Springer TA. Structural specializations of A2, a force-sensing domain in the ultralarge vascular protein von Willebrand factor. Proc Natl Acad Sci U S A. 2009;106:9226–9231. doi: 10.1073/pnas.0903679106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Zhang X, Halvorsen K, Zhang C-Z, Wong WP, Springer TA. Mechanoenzymatic cleavage of the ultralarge vascular protein von Willebrand factor. Science. 2009;324:1330–1334. doi: 10.1126/science.1170905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Nesbitt WS, Kulkarni S, Giuliano S, Goncalves I, Dopheide SM, Yap CL, Harper IS, Salem HH, Jackson SP. Distinct glycoprotein Ib/V/IX and integrin αIIbβ3-dependent calcium signals cooperatively regulate platelet adhesion under flow. J Biol Chem. 2002;277:2965–2972. doi: 10.1074/jbc.M110070200. [DOI] [PubMed] [Google Scholar]
  • 8.Yap CL, Anderson KE, Hughan SC, Dopheide SM, Salem HH, Jackson SP. Essential role for phosphoinositide 3-kinase in shear-dependent signaling between platelet glycoprotein Ib/V/IX and integrin αIIbβ3. Blood. 2002;99:151–158. doi: 10.1182/blood.v99.1.151. [DOI] [PubMed] [Google Scholar]
  • 9.Mazzucato M, Pradella P, Cozzi MR, De Marco L, Ruggeri ZM. Sequential cytoplasmic calcium signals in a 2-stage platelet activation process induced by the glycoprotein Ibα mechanoreceptor. Blood. 2002;100:2793–2800. doi: 10.1182/blood-2002-02-0514. [DOI] [PubMed] [Google Scholar]
  • 10.Goncalves I, Nesbitt WS, Yuan Y, Jackson SP. Importance of temporal flow gradients and integrin αIIbβ3 mechanotransduction for shear activation of platelets. J Biol Chem. 2005;280:15430–15437. doi: 10.1074/jbc.M410235200. [DOI] [PubMed] [Google Scholar]
  • 11.Mazzucato M, Cozzi MR, Pradella P, Ruggeri ZM, De Marco L. Distinct roles of ADP receptors in von Willebrand factor-mediated platelet signaling and activation under high flow. Blood. 2004;104:3221–3227. doi: 10.1182/blood-2004-03-1145. [DOI] [PubMed] [Google Scholar]
  • 12.Feng S, Lu X, Reséndiz JC, Kroll MH. Pathological shear stress directly regulates platelet αIIbβ3 signaling. Am J Physiol Cell Physiol. 2006;291:C1346–C1354. doi: 10.1152/ajpcell.00559.2005. [DOI] [PubMed] [Google Scholar]
  • 13.Jackson SP. The growing complexity of platelet aggregation. Blood. 2007;109:5087–5095. doi: 10.1182/blood-2006-12-027698. [DOI] [PubMed] [Google Scholar]
  • 14.Pozgajová M, Sachs UJH, Hein L, Nieswandt B. Reduced thrombus stability in mice lacking the α2A-adrenergic receptor. Blood. 2006;108:510–514. doi: 10.1182/blood-2005-12-4835. [DOI] [PubMed] [Google Scholar]
  • 15.Andre P, Delaney SM, LaRocca T, Vincent D, DeGuzman F, Jurek M, Koller B, Phillips DR, Conley PB. P2Y12 regulates platelet adhesion/activation, thrombus growth, and thrombus stability in injured arteries. J Clin Invest. 2003;112:398–406. doi: 10.1172/JCI17864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Prévost N, Woulfe DS, Jiang H, Stalker TJ, Marchese P, Ruggeri ZM, Brass LF. Eph kinases and ephrins support thrombus growth and stability by regulating integrin outside-in signaling in platelets. Proc Natl Acad Sci USA. 2005;102:9820–9825. doi: 10.1073/pnas.0404065102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Goto S, Tamura N, Ishida H, Ruggeri ZM. Dependence of platelet thrombus stability on sustained glycoprotein IIb/IIIa activation through adenosine 5'-diphosphate receptor stimulation and cyclic calcium signaling. J Am Coll Cardiol. 2006;47:155–162. doi: 10.1016/j.jacc.2005.08.055. [DOI] [PubMed] [Google Scholar]
  • 18.Speich HE, Earhart AD, Hill SN, Cholera S, Kueter TJ, Smith JN, White MM, Jennings LK. Variability of platelet aggregate dispersal with glycoprotein IIb-IIIa antagonists eptifibatide and abciximab. J Thromb Haemost. 2009;7:983–991. doi: 10.1111/j.1538-7836.2009.03432.x. [DOI] [PubMed] [Google Scholar]
  • 19.Speich HE, Earhart AD, Hill SN, Kueter TJ, White MM, Jennings LK. Platelet aggregation reversal by GPIIb-IIIa antagonists: dependence upon agonist and aggregate age. Thromb Haemost. 2009;7:PP-TH-097. doi: 10.1111/j.1538-7836.2009.03432.x. [DOI] [PubMed] [Google Scholar]
  • 20.Deibele AJ, Jennings LK, Tcheng JE, Neva C, Earhart AD, Gibson CM. Intracoronary eptifibatide bolus administration during percutaneous coronary revascularization for acute coronary syndromes with evaluation of platelet glycoprotein IIb/IIIa receptor occupancy and platelet function: the IntraCoronary Eptifibatide (ICE) Trial. Circulation. 2010;121:784–791. doi: 10.1161/CIRCULATIONAHA.109.882746. [DOI] [PubMed] [Google Scholar]
  • 21.Huo Y, Ley KF. Role of platelets in the development of atherosclerosis. Trends Cardiovasc Med. 2004;14:18–22. doi: 10.1016/j.tcm.2003.09.007. [DOI] [PubMed] [Google Scholar]
  • 22.Linden MD, Jackson DE. Platelets: pleiotropic roles in atherogenesis and atherothrombosis. Int J Biochem Cell Biol. 2010;42:1762–1766. doi: 10.1016/j.biocel.2010.07.012. [DOI] [PubMed] [Google Scholar]
  • 23.Cyrus T, Yao Y, Ding T, Dogné JM, Praticò D. Thromboxane receptor blockade improves the antiatherogenic effect of thromboxane A2 suppression in LDLR KO mice. Blood. 2007;109:3291–3296. doi: 10.1182/blood-2006-08-044990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Methia N, André P, Denis CV, Economopoulos M, Wagner DD. Localized reduction of atherosclerosis in von Willebrand factor-deficient mice. Blood. 2001;98:1424–1428. doi: 10.1182/blood.v98.5.1424. [DOI] [PubMed] [Google Scholar]
  • 25.Massberg S, Brand K, Grüner S, Page S, Müller E, Müller I, Bergmeier W, Richter T, Lorenz M, Konrad I, Nieswandt B, Gawaz M. A critical role of platelet adhesion in the initiation of atherosclerotic lesion formation. J Exp Med. 2002;196:887–896. doi: 10.1084/jem.20012044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.McCarty OJT, Conley RB, Shentu W, Tormoen GW, Zha D, Xie A, Qi Y, Zhao Y, Carr C, Belcik T, Keene DR, de Groot PG, Lindner JR. Molecular imaging of activated von Willebrand factor to detect high-risk atherosclerotic phenotype. J Am Coll Cardiol Cardiovasc Imaging. 2010;3:947–955. doi: 10.1016/j.jcmg.2010.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Fu X, Chen J, Gallagher R, Zheng Y, Chung DW, López JA. Shear stress-induced unfolding of VWF accelerates oxidation of key methionine residues in the A1A2A3 region. Blood. 2011;118:5283–5291. doi: 10.1182/blood-2011-01-331074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hu H, Varon D, Hjemdahl P, Savion N, Schulman S, Li N. Platelet-leukocyte aggregation under shear stress: differential involvement of selectins and integrins. Thromb Haemost. 2003;90:679–687. doi: 10.1160/TH03-05-0274. [DOI] [PubMed] [Google Scholar]
  • 29.Ridger V, Krams R, Carpi A, Evans PC. Hemodynamic parameters regulating vascular inflammation and atherosclerosis: a brief update. Biomed Pharmacother. 2008;62:536–540. doi: 10.1016/j.biopha.2008.07.053. [DOI] [PubMed] [Google Scholar]
  • 30.Gurbel PA, Becker RC, Mann KG, Steinhubl SR, Michelson AD. Platelet function monitoring in patients with coronary artery disease. J Am Coll Cardiol. 2007;50:1822–1834. doi: 10.1016/j.jacc.2007.07.051. [DOI] [PubMed] [Google Scholar]
  • 31.Falati S, Gross P, Merrill-Skoloff G, Furie BC, Furie B. Real-time in vivo imaging of platelets, tissue factor and fibrin during arterial thrombus formation in the mouse. Nat Med. 2002;8:1175–1181. doi: 10.1038/nm782. [DOI] [PubMed] [Google Scholar]
  • 32.Gross PL, Ni R, Esposito B, Rand ML, Weitz JI. The dynamics of platelet activation in vivo. J Thromb Haemost. 2009;7:PP-TH-009. [Google Scholar]
  • 33.Yong ASC, Pennings GJ, Chang M, Hamzah A, Chung T, Qi M, Brieger D, Behnia M, Krilis SA, Ng MKC, Lowe HC, Kritharides L. Intra-coronary shear-related up-regulation of platelet P-selectin and platelet-monocyte aggregation despite the use of aspirin and clopidogrel. Blood. 2011;117:11–20. doi: 10.1182/blood-2010-04-278812. [DOI] [PubMed] [Google Scholar]
  • 34.Hellums JD, Peterson DM, Stathopoulos NA, Moake JL, Giorgio TD. Cerebral Ischemia and Hemorheology. Springer-Verlag; New York: 1987. Studies on the mechanisms of shear-induced platelet activation; pp. 80–89. [Google Scholar]
  • 35.Kolandaivelu K, Swaminathan R, Gibson WJ, Kolachalama VB, Nguyen-Ehrenreich K-L, Giddings VL, Coleman L, Wong GK, Edelman ER. Stent thrombogenicity early in high-risk interventional settings is driven by stent design and deployment and protected by polymer-drug coatings. Circulation. 2011;123:1400–1409. doi: 10.1161/CIRCULATIONAHA.110.003210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Yin W, Alemu Y, Affeld K, Jesty J, Bluestein D. Flow-induced platelet activation in bileaflet and monoleaflet mechanical heart valves. Ann Biomed Eng. 2004;32:1058–1066. doi: 10.1114/b:abme.0000036642.21895.3f. [DOI] [PubMed] [Google Scholar]
  • 37.Yin W, Krukenkamp IB, Saltman AE, Gaudette G, Suresh K, Bernal O, Jesty J, Bluestein D. Thrombogenic performance of a St Jude bileaflet mechanical heart valve in a sheep model. ASAIO J. 2006;52:28–33. doi: 10.1097/01.mat.0000198123.42686.ca. [DOI] [PubMed] [Google Scholar]
  • 38.Nobili M, Morbiducci U, Ponzini R, Del Gaudio C, Balducci A, Grigioni M, Maria Montevecchi F, Redaelli A. Numerical simulation of the dynamics of a bileaflet prosthetic heart valve using a fluid-structure interaction approach. J Biomech. 2008;41:2539–2550. doi: 10.1016/j.jbiomech.2008.05.004. [DOI] [PubMed] [Google Scholar]
  • 39.Bluestein D, Chandran KB, Manning KB. Towards non-thrombogenic performance of blood recirculating devices. Ann Biomed Eng. 2010;38:1236–1256. doi: 10.1007/s10439-010-9905-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Alemu Y, Girdhar G, Xenos M, Sheriff J, Jesty J, Einav S, Bluestein D. Design optimization of a mechanical heart valve for reducing valve thrombogenicity: a case study with ATS valve. ASAIO J. 2010;56:389–396. doi: 10.1097/MAT.0b013e3181e65bf9. [DOI] [PubMed] [Google Scholar]
  • 41.Xenos M, Girdhar G, Alemu Y, Jesty J, Slepian M, Einav S, Bluestein D. Device thrombogenicity emulator (DTE): design optimization methodology for cardiovascular devices: a study in two bileaflet MHV designs. J Biomech. 2010;43:2400–2409. doi: 10.1016/j.jbiomech.2010.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Girdhar G, Bluestein D. Biological effects of dynamic shear stress in cardiovascular pathologies and devices. Expert Rev Med Devices. 2008;5:167–181. doi: 10.1586/17434440.5.2.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Palmerini T, Nedelman MA, Scudder LE, Nakada MT, Jordan RE, Smyth S, Gordon RE, Fallon JT, Coller BS. Effects of abciximab on the acute pathology of blood vessels after arterial stenting in nonhuman primates. J Am Coll Cardiol. 2002;40:360–366. doi: 10.1016/s0735-1097(02)01951-4. [DOI] [PubMed] [Google Scholar]
  • 44.Rowley JW, Oler A, Tolley ND, Hunter B, Low EN, Nix DA, Yost CC, Zimmerman GA, Weyrich AS. Genome wide RNA-seq analysis of human and mouse platelet transcriptomes. Blood. 2011;118:e101–e111. doi: 10.1182/blood-2011-03-339705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Landry P, Plante I, Ouellet DL, Perron MP, Rousseau G, Provost P. Existence of a microRNA pathway in anucleate platelets. Nat Struct Mol Biol. 2009;16:961–966. doi: 10.1038/nsmb.1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Zimmerman GA, Weyrich AS. Signal-dependent protein synthesis by activated platelets: new pathways to altered phenotype and function. Arterioscler Thromb Vasc Biol. 2008;28:s17–s24. doi: 10.1161/ATVBAHA.107.160218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Freedman JE, Larson MG, Tanriverdi K, O'Donnell CJ, Morin K, Hakanson AS, Vasan RS, Johnson AD, Iafrati MD, Benjamin EJ. Relation of platelet and leukocyte inflammatory transcripts to body mass index in the Framingham Heart Study. Circulation. 2010;122:119–129. doi: 10.1161/CIRCULATIONAHA.109.928192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Willerson JT, Yao SK, McNatt J, Benedict CR, Anderson HV, Golino P, Murphree SS, Buja LM. Frequency and severity of cyclic flow alternations and platelet aggregation predict the severity of neointimal proliferation following experimental coronary stenosis and endothelial injury. Proc Natl Acad Sci U S A. 1991;88:10624–10628. doi: 10.1073/pnas.88.23.10624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Anderson HV, McNatt J, Clubb FJ, Herman M, Maffrand JP, DeClerck F, Ahn C, Buja LM, Willerson JT. Platelet inhibition reduces cyclic flow variations and neointimal proliferation in normal and hypercholesterolemic-atherosclerotic canine coronary arteries. Circulation. 2001;104:2331–2337. doi: 10.1161/hc4401.098434. [DOI] [PubMed] [Google Scholar]
  • 50.Bienvenu JG, Tanguay JF, Théorêt JF, Kumar A, Schaub RG, Merhi Y. Recombinant soluble P-selectin glycoprotein ligand-1-Ig reduces restenosis through inhibition of platelet-neutrophil adhesion after double angioplasty in swine. Circulation. 2001;103:1128–1134. doi: 10.1161/01.cir.103.8.1128. [DOI] [PubMed] [Google Scholar]
  • 51.Kabbani SS, Watkins MW, Ashikaga T, Terrien EF, Holoch PA, Sobel BE, Schneider DJ. Platelet reactivity characterized prospectively: a determinant of outcome 90 days after percutaneous coronary intervention. Circulation. 2001;104:181–186. doi: 10.1161/01.cir.104.2.181. [DOI] [PubMed] [Google Scholar]
  • 52.Steinhubl SR, Saucedo JF, Sane DC, Berger PB, Moliterno DJ, Tcheng JE, Kereiakes DJ. Point-of-care measurement of platelet function before angioplasty strongly predicts future target vessel revascularization. J Am Coll Cardiol. 2002;37:72A. [Google Scholar]
  • 53.Bhatt DL, Fox KAA, Hacke W, Berger PB, Black HR, Boden WE, Cacoub P, Cohen EA, Creager MA, Easton JD, Flather MD, Haffner SM, Hamm CW, Hankey GJ, Johnston SC, Mak K-H, Mas J-L, Montalescot G, Pearson TA, Steg PG, Steinhubl SR, Weber MA, Brennan DM, Fabry-Ribaudo L, Booth J, Topol EJ. Clopidogrel and aspirin versus aspirin alone for the prevention of atherothrombotic events. N Engl J Med. 2006;354:1706–1717. doi: 10.1056/NEJMoa060989. [DOI] [PubMed] [Google Scholar]
  • 54.Schwartz L, Bourassa MG, Lespérance J, Aldridge HE, Kazim F, Salvatori VA, Henderson M, Bonan R, David PR. Aspirin and dipyridamole in the prevention of restenosis after percutaneous transluminal coronary angioplasty. N Engl J Med. 1988;318:1714–1719. doi: 10.1056/NEJM198806303182603. [DOI] [PubMed] [Google Scholar]
  • 55.Steinhubl SR, Berger PB, Mann JT, III, Fry ETA, DeLago A, Wilmer C, Topol EJ. Early and sustained dual oral antiplatelet therapy following percutaneous coronary intervention: a randomized controlled trial. JAMA. 2002;288:2411–2420. doi: 10.1001/jama.288.19.2411. [DOI] [PubMed] [Google Scholar]
  • 56.Topol EJ, Califf RM, Weisman HF, Ellis SG, Tcheng JE, Worley S, Ivanhoe R, George BS, Fintel D, Weston M, EPIC Investigators Randomised trial of coronary intervention with antibody against platelet IIb/IIIa integrin for reduction of clinical restenosis: results at six months. Lancet. 1994;343:881–886. doi: 10.1016/s0140-6736(94)90007-8. [DOI] [PubMed] [Google Scholar]
  • 57.EPISTENT Investigators Randomised placebo-controlled and balloon-angioplasty-controlled trial to assess safety of coronary stenting with use of platelet glycoprotein-IIb/IIIa blockade. Lancet. 1998;352:87–92. doi: 10.1016/s0140-6736(98)06113-3. [DOI] [PubMed] [Google Scholar]
  • 58.Neumann FJ, Kastrati A, Schmitt C, Blasini R, Hadamitzky M, Mehilli J, Gawaz M, Schleef M, Seyfarth M, Dirschinger J, Schömig A. Effect of glycoprotein IIb/IIIa receptor blockade with abciximab on clinical and angiographic restenosis rate after the placement of coronary stents following acute myocardial infarction. J Am Coll Cardiol. 2000;35:915–921. doi: 10.1016/s0735-1097(99)00635-x. [DOI] [PubMed] [Google Scholar]
  • 59.ERASER Investigators Acute platelet inhibition with abciximab does not reduce in-stent restenosis (ERASER study) Circulation. 1999;100:799–806. doi: 10.1161/01.cir.100.8.799. [DOI] [PubMed] [Google Scholar]
  • 60.Montalescot G, Barragan P, Wittenberg O, Ecollan P, Elhadad S, Villain P, Boulenc JM, Morice MC, Maillard L, Pansiéri M, Choussat R, Pinton P. Platelet glycoprotein IIb/IIIa inhibition with coronary stenting for acute myocardial infarction. N Engl J Med. 2001;344:1895–1903. doi: 10.1056/NEJM200106213442503. [DOI] [PubMed] [Google Scholar]
  • 61.Tamhane U, Meier P, Chetcuti S, Chen K-Y, Rha S-W, Grossman MP, Gurm H. Efficacy of cilostazol in reducing restenosis in patients undergoing contemporary stent based PCI: a meta-analysis of randomised controlled trials. EuroIntervention. 2009;5:384–393. doi: 10.4244/v5i3a60. [DOI] [PubMed] [Google Scholar]
  • 62.Lee CW, Park D-W, Seung KB, Kim PJ, Park HJ, Kim W-J, Lee JY, Kang S-J, Lee S-H, Kim Y-H, Park S-W, Park S-J. Comparison of dual drug-eluting Cilotax stent and paclitaxel-eluting Taxus Liberté stent in native coronary artery lesions. Am J Cardiol. 2011;107:990–994. doi: 10.1016/j.amjcard.2010.11.021. [DOI] [PubMed] [Google Scholar]
  • 63.Costa MA, Simon DI. Molecular basis of restenosis and drug-eluting stents. Circulation. 2005;111:2257–2273. doi: 10.1161/01.CIR.0000163587.36485.A7. [DOI] [PubMed] [Google Scholar]
  • 64.Sanguigni V, Pignatelli P, Lenti L, Ferro D, Bellia A, Carnevale R, Tesauro M, Sorge R, Lauro R, Violi F. Short-term treatment with atorvastatin reduces platelet CD40 ligand and thrombin generation in hypercholesterolemic patients. Circulation. 2005;111:412–419. doi: 10.1161/01.CIR.0000153810.81187.7D. [DOI] [PubMed] [Google Scholar]
  • 65.Nissen SE, Nicholls SJ, Sipahi I, Libby P, Raichlen JS, Ballantyne CM, Davignon J, Erbel R, Fruchart JC, Tardif J-C, Schoenhagen P, Crowe T, Cain V, Wolski K, Goormastic M, Tuzcu EM. Effect of very high-intensity statin therapy on regression of coronary atherosclerosis: the ASTEROID trial. JAMA. 2006;295:1556–1565. doi: 10.1001/jama.295.13.jpc60002. [DOI] [PubMed] [Google Scholar]
  • 66.Nissen SE, Tuzcu EM, Schoenhagen P, Brown BG, Ganz P, Vogel RA, Crowe T, Howard G, Cooper CJ, Brodie B, Grines CL, DeMaria AN. Effect of intensive compared with moderate lipid-lowering therapy on progression of coronary atherosclerosis: a randomized controlled trial. JAMA. 2004;291:1071–1080. doi: 10.1001/jama.291.9.1071. [DOI] [PubMed] [Google Scholar]
  • 67.Lievens D, Zernecke A, Seijkens T, Soehnlein O, Beckers L, Munnix ICA, Wijnands E, Goossens P, van Kruchten R, Thevissen L, Boon L, Flavell RA, Noelle RJ, Gerdes N, Biessen EA, Daemen MJAP, Heemskerk JWM, Weber C, Lutgens E. Platelet CD40L mediates thrombotic and inflammatory processes in atherosclerosis. Blood. 2010;116:4317–4327. doi: 10.1182/blood-2010-01-261206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Pasceri V, Patti G, Nusca A, Pristipino C, Richichi G, Di Sciascio G. Randomized trial of atorvastatin for reduction of myocardial damage during coronary intervention: results from the ARMYDA (Atorvastatin for Reduction of MYocardial Damage during Angioplasty) study. Circulation. 2004;110:674–678. doi: 10.1161/01.CIR.0000137828.06205.87. [DOI] [PubMed] [Google Scholar]
  • 69.Briguori C, Visconti G, Focaccio A, Golia B, Chieffo A, Castelli A, Mussardo M, Montorfano M, Ricciardelli B, Colombo A. Novel Approaches for Preventing or Limiting EventS (NAPLES) II trial: impact of a single high loading dose of atorvastatin on periprocedural myocardial infarction. J Am Coll Cardiol. 2009;54:2157–2163. doi: 10.1016/j.jacc.2009.07.005. [DOI] [PubMed] [Google Scholar]
  • 70.Puccetti L, Santilli F, Pasqui AL, Lattanzio S, Liani R, Ciani F, Ferrante E, Ciabattoni G, Scarpini F, Ghezzi A, Auteri A, Davì G. Effects of atorvastatin and rosuvastatin on thromboxane-dependent platelet activation and oxidative stress in hypercholesterolemia. Atherosclerosis. 2011;214:122–128. doi: 10.1016/j.atherosclerosis.2010.10.006. [DOI] [PubMed] [Google Scholar]
  • 71.Matetzky S, Fefer P, Shenkman B, Shechter M, Novikov I, Savion N, Varon D, Hod H. Statins have an early antiplatelet effect in patients with acute myocardial infarction. Platelets. 2011;22:103–110. doi: 10.3109/09537104.2010.512402. [DOI] [PubMed] [Google Scholar]
  • 72.van der Loo B, Spring S, Koppensteiner R. High-dose atorvastatin treatment in patients with peripheral arterial disease: effects on platelet aggregation, blood rheology and plasma homocysteine. Clin Hemorheol Microcirc. 2011;47:241–251. doi: 10.3233/CH-2011-1386. [DOI] [PubMed] [Google Scholar]
  • 73.Schwartz RS, Holmes DR, Jr, Topol EJ. The restenosis paradigm revisited: an alternative proposal for cellular mechanisms. J Am Coll Cardiol. 1992;20:1284–1293. doi: 10.1016/0735-1097(92)90389-5. [DOI] [PubMed] [Google Scholar]
  • 74.Wang K, Zhou Z, Zhou X, Tarakji K, Topol EJ, Lincoff AM. Prevention of intimal hyperplasia with recombinant soluble P-selectin glycoprotein ligand-immunoglobulin in the porcine coronary artery balloon injury model. J Am Coll Cardiol. 2001;38:577–582. doi: 10.1016/s0735-1097(01)01347-x. [DOI] [PubMed] [Google Scholar]
  • 75.Hayashi S, Watanabe N, Nakazawa K, Suzuki J, Tsushima K, Tamatani T, Sakamoto S, Isobe M. Roles of P-selectin in inflammation, neointimal formation, and vascular remodeling in balloon-injured rat carotid arteries. Circulation. 2000;102:1710–1717. doi: 10.1161/01.cir.102.14.1710. [DOI] [PubMed] [Google Scholar]
  • 76.Rogers C, Welt FG, Karnovsky MJ, Edelman ER. Monocyte recruitment and neointimal hyperplasia in rabbits: coupled inhibitory effects of heparin. Arterioscler Thromb Vasc Biol. 1996;16:1312–1318. doi: 10.1161/01.atv.16.10.1312. [DOI] [PubMed] [Google Scholar]
  • 77.Pache J, Kastrati A, Mehilli J, Schühlen H, Dotzer F, Hausleiter J, Fleckenstein M, Neumann FJ, Sattelberger U, Schmitt C, Müller M, Dirschinger J, Schömig A. Intracoronary stenting and angiographic results: strut thickness effect on restenosis outcome (ISAR-STEREO-2) trial. J Am Coll Cardiol. 2003;41:1283–1288. doi: 10.1016/s0735-1097(03)00119-0. [DOI] [PubMed] [Google Scholar]
  • 78.Rittersma SZH, de Winter RJ, Koch KT, Bax M, Schotborgh CE, Mulder KJ, Tijssen JGP, Piek JJ. Impact of strut thickness on late luminal loss after coronary artery stent placement. Am J Cardiol. 2004;93:477–480. doi: 10.1016/j.amjcard.2003.10.049. [DOI] [PubMed] [Google Scholar]
  • 79.Turco MA, Ormiston JA, Popma JJ, Hall JJ, Mann T, Cannon LA, Webster MWI, Mishkel GJ, O'Shaughnessy CD, McGarry TF, Mandinov L, Dawkins KD, Baim DS. Reduced risk of restenosis in small vessels and reduced risk of myocardial infarction in long lesions with the new thin-strut TAXUS Liberté stent: 1-year results from the TAXUS ATLAS program. J Am Coll Cardiol Cardiovasc Interv. 2008;1:699–709. doi: 10.1016/j.jcin.2008.09.007. [DOI] [PubMed] [Google Scholar]
  • 80.Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, Horrow J, Husted S, James S, Katus H, Mahaffey KW, Scirica BM, Skene A, Steg PG, Storey RF, Harrington RA, Freij A, Thorsén M. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2009;361:1045–1057. doi: 10.1056/NEJMoa0904327. [DOI] [PubMed] [Google Scholar]
  • 81.Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Gottlieb S, Neumann F-J, Ardissino D, De Servi S, Murphy SA, Riesmeyer J, Weerakkody G, Gibson CM, Antman EM. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007;357:2001–2015. doi: 10.1056/NEJMoa0706482. [DOI] [PubMed] [Google Scholar]
  • 82.Joner M, Finn AV, Farb A, Mont EK, Kolodgie FD, Ladich E, Kutys R, Skorija K, Gold HK, Virmani R. Pathology of drug-eluting stents in humans: delayed healing and late thrombotic risk. J Am Coll Cardiol. 2006;48:193–202. doi: 10.1016/j.jacc.2006.03.042. [DOI] [PubMed] [Google Scholar]
  • 83.Lagerqvist B, Carlsson J, Fröbert O, Lindbäck J, Scherstén F, Stenestrand U, James SK. Stent thrombosis in Sweden: a report from the Swedish Coronary Angiography and Angioplasty Registry. Circ Cardiovasc Interv. 2009;2:401–408. doi: 10.1161/CIRCINTERVENTIONS.108.844985. [DOI] [PubMed] [Google Scholar]
  • 84.Finn AV, Joner M, Nakazawa G, Kolodgie F, Newell J, John MC, Gold HK, Virmani R. Pathological correlates of late drug-eluting stent thrombosis: strut coverage as a marker of endothelialization. Circulation. 2007;115:2435–2441. doi: 10.1161/CIRCULATIONAHA.107.693739. [DOI] [PubMed] [Google Scholar]
  • 85.Nebeker JR, Virmani R, Bennett CL, Hoffman JM, Samore MH, Alvarez J, Davidson CJ, McKoy JM, Raisch DW, Whisenant BK, Yarnold PR, Belknap SM, West DP, Gage JE, Morse RE, Gligoric G, Davidson L, Feldman MD. Hypersensitivity cases associated with drug-eluting coronary stents: a review of available cases from the Research on Adverse Drug Events and Reports (RADAR) project. J Am Coll Cardiol. 2006;47:175–181. doi: 10.1016/j.jacc.2005.07.071. [DOI] [PubMed] [Google Scholar]
  • 86.Nakazawa G, Finn AV, Vorpahl M, Ladich ER, Kolodgie FD, Virmani R. Coronary responses and differential mechanisms of late stent thrombosis attributed to first-generation sirolimus- and paclitaxel-eluting stents. JAm Coll Cardiol. 2011;57:390–398. doi: 10.1016/j.jacc.2010.05.066. [DOI] [PubMed] [Google Scholar]
  • 87.Nakazawa G, Finn AV, Joner M, Ladich E, Kutys R, Mont EK, Gold HK, Burke AP, Kolodgie FD, Virmani R. Delayed arterial healing and increased late stent thrombosis at culprit sites after drug-eluting stent placement for acute myocardial infarction patients: an autopsy study. Circulation. 2008;118:1138–1145. doi: 10.1161/CIRCULATIONAHA.107.762047. [DOI] [PubMed] [Google Scholar]
  • 88.Nakazawa G, Otsuka F, Nakano M, Vorpahl M, Yazdani SK, Ladich E, Kolodgie FD, Finn AV, Virmani R. The pathology of neoatherosclerosis in human coronary implants bare-metal and drug-eluting stents. J Am Coll Cardiol. 2011;57:1314–1322. doi: 10.1016/j.jacc.2011.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Kwon SW, Kim B-K, Kim T-H, Kim J-S, Ko Y-G, Choi D, Jang Y, Hong M-K. Qualitative assessment of neointimal tissue after drug-eluting stent implantation: comparison between follow-up optical coherence tomography and intravascular ultrasound. Am Heart J. 2011;161:367–372. doi: 10.1016/j.ahj.2010.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Murata A, Wallace-Bradley D, Tellez A, Alviar C, Aboodi M, Sheehy A, Coleman L, Perkins L, Nakazawa G, Mintz G, Kaluza GL, Virmani R, Granada JF. Accuracy of optical coherence tomography in the evaluation of neointimal coverage after stent implantation. J Am Coll Cardiol Cardiovasc Imaging. 2010;3:76–84. doi: 10.1016/j.jcmg.2009.09.018. [DOI] [PubMed] [Google Scholar]
  • 91.Morlacchi S, Keller B, Arcangeli P, Balzan M, Migliavacca F, Dubini G, Gunn J, Arnold N, Narracott A, Evans D, Lawford P. Hemodynamics and in-stent restenosis: micro-CT images, histology, and computer simulations. Ann Biomed Eng. 2011;39:2615–2626. doi: 10.1007/s10439-011-0355-9. [DOI] [PubMed] [Google Scholar]
  • 92.Hu Z-Y, Chen S-L, Zhang J-J, Shan S-J, Liu Z-Z, Ye F, Kan J, Xu H-M, Nguyen K, Kwan T, Nguyen T, Hoang T. Distribution and magnitude of shear stress after coronary bifurcation lesions stenting with the classical crush technique: a new predictor for in-stent restenosis. J Interv Cardiol. 2010;23:330–340. doi: 10.1111/j.1540-8183.2010.00571.x. [DOI] [PubMed] [Google Scholar]
  • 93.Papafaklis MI, Bourantas CV, Theodorakis PE, Katsouras CS, Naka KK, Fotiadis DI, Michalis LK. The effect of shear stress on neointimal response following sirolimus- and paclitaxel-eluting stent implantation compared with bare-metal stents in humans. J Am Coll Cardiol Cardiovasc Interv. 2010;3:1181–1189. doi: 10.1016/j.jcin.2010.08.018. [DOI] [PubMed] [Google Scholar]
  • 94.Baber U, Mehran R, Sharma SK, Brar S, Yu J, Suh J-W, Kim H-S, Park S-J, Kastrati A, de Waha A, Krishnan P, Moreno P, Sweeny J, Kim MC, Suleman J, Pyo R, Wiley J, Kovacic J, Kini AS, Dangas GD. Impact of the everolimus-eluting stent on stent thrombosis: a meta-analysis of 13 randomized trials. J Am Coll Cardiol. 2011;58:1569–1577. doi: 10.1016/j.jacc.2011.06.049. [DOI] [PubMed] [Google Scholar]
  • 95.Park KW, Chae I-H, Lim D-S, Han K-R, Yang H-M, Lee H-Y, Kang H-J, Koo B-K, Ahn T, Yoon J-H, Jeong M-H, Hong T-J, Chung W-Y, Jo S-H, Choi Y-J, Hur S-H, Kwon H-M, Jeon D-W, Kim B-O, Park S-H, Lee N-H, Jeon H-K, Gwon H-C, Jang Y-S, Kim H-S. Everolimus-eluting versus sirolimus-eluting stents in patients undergoing percutaneous coronary intervention: the EXCELLENT (Efficacy of Xience/Promus Versus Cypher to Reduce Late Loss After Stenting) randomized trial. J Am Coll Cardiol. 2011;58:1844–1854. doi: 10.1016/j.jacc.2011.07.031. [DOI] [PubMed] [Google Scholar]
  • 96.Dauerman HL. The magic of disappearing stents. J Am Coll Cardiol. 2011;58:1589–1591. doi: 10.1016/j.jacc.2011.05.049. [DOI] [PubMed] [Google Scholar]

RESOURCES