Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Pre PCI hospital antithrombotic therapy for ST elevation myocardial infarction: striving for consensus S. Michael Gharacholou, Brenda J. Larson, Christian C. Zuver, Ryan J. Wubben, Giorgio Gimelli & Amish N. Raval Journal of Thrombosis and Thrombolysis A Journal for Translation, Application and Therapeutics in Thrombosis and Vascular Science ISSN 0929-5305 J Thromb Thrombolysis DOI 10.1007/s11239-012-0744-4 1 23 Your article is protected by copyright and all rights are held exclusively by Springer Science+Business Media, LLC. This e-offprint is for personal use only and shall not be selfarchived in electronic repositories. If you wish to self-archive your work, please use the accepted author’s version for posting to your own website or your institution’s repository. You may further deposit the accepted author’s version on a funder’s repository at a funder’s request, provided it is not made publicly available until 12 months after publication. 1 23 Author's personal copy J Thromb Thrombolysis DOI 10.1007/s11239-012-0744-4 Pre PCI hospital antithrombotic therapy for ST elevation myocardial infarction: striving for consensus S. Michael Gharacholou • Brenda J. Larson Christian C. Zuver • Ryan J. Wubben • Giorgio Gimelli • Amish N. Raval • Ó Springer Science+Business Media, LLC 2012 Abstract Strong evidence exists in favor of rapid transfer of a patient suffering an ST-elevation myocardial infarction (STEMI) to the nearest hospital with primary percutaneous coronary intervention (PCI) capability, assuming the time from first medical contact to balloon inflation can be achieved in less than 90 min. In many areas, PCI hospitals have successfully collaborated with regional non-PCI hospitals to provide primary PCI for STEMI; however, significant variations exist in how these programs are executed. For example, the pre PCI hospital administration of antithrombotic agents by emergency medical personnel can include aspirin, clopidogrel, unfractionated heparin, low molecular weight heparin, partial or full dose fibrinolytics or combinations thereof. There is little consensus on the optimal cocktail, dose and route of administration. Standardizing the pre PCI antithrombotic regimen across hospital systems may be one approach to improve timely administration of these therapies, and potentially improve STEMI outcomes. S. M. Gharacholou (&) B. J. Larson G. Gimelli A. N. Raval Division of Cardiovascular Medicine, University of Wisconsin Hospital and Clinics, 600 Highland Avenue, Madison, WI 53792, USA e-mail: [email protected] C. C. Zuver R. J. Wubben Division of Emergency Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA Introduction Half a million people suffer ST-elevation myocardial infarction (STEMI) every year. Rapidly restoring blood flow in the occluded culprit coronary artery offers the best opportunity to preserve ventricular function, prevent heart failure and reduce mortality [1]. Primary percutaneous coronary intervention (PCI) is preferred over fibrinolysis provided that experienced operators can implement this treatment within 90 min from first medical contact. However, approximately 75 % of STEMI patients (roughly 350,000 patients per year) initially present to health care facilities incapable of performing primary PCI [2]. Furthermore, it is estimated that between 7 and 30 % of patients fail to receive lifesaving fibrinolytic therapy or primary PCI, despite having no contraindications [3, 4]. One-fifth of STEMI patients have contraindications to fibrinolytics and 70 % of these fail to receive timely primary PCI [2, 5]. These sobering statistics have reinforced the importance of standardized, system-wide delivery of evidence-based STEMI care. The ‘‘hub and spoke’’ care model for STEMI has recently emerged in many regions of the US. This structure allows hospitals without onsite PCI capabilities (spokes) to coordinate patient care with PCI hospitals (hubs) [6, 7]. The American Heart Association (AHA), in collaboration with emergency medicine and third-party insurers, has advocated for collaborative regional system approaches for STEMI management [2]. The role of emergency medical service (EMS) in any effective regional STEMI program is multifaceted and includes: recognition of probable STEMI, rapid transport to the most appropriate PCI capable facility, and administration of active therapy en route [1, 8, 9]. EMS transport of STEMI patients from the scene to a PCI hospital has been associated with improved outcomes, even 123 Author's personal copy S. M. Gharacholou et al. among patients who are administered fibrinolysis and are transferred for PCI within 6 h of fibrinolysis [10]. Furthermore, there is interest in EMS-directed initiatives to transport STEMI patients directly to centers capable of performing PCI, rather than invoking interhospital transfer from regional non-PCI hospitals; however, no controlled trials testing this strategy have been performed. Generally, paramedic units responding to calls for probable STEMI are likely to initiate preparatory antithrombotic agents; however, variations exist in the type of medications, doses and routes of administration. A principle goal in regional STEMI programs is to increase the proportion of STEMI patients who receive timely and successful reperfusion; however, a unified perspective on the optimal pre PCI hospital antithrombotic regimen has not been offered. Herein, we will review contemporary antiplatelet and antithrombotic therapy in patients with STEMI referred for PCI. We will explore the potential time-dependent benefits of agents administered in the pre-hospital setting and highlight newer antithrombotic agents and their potential implications for STEMI management. Finally, we will share the rationale behind our own regional, standardized approach to pre PCI hospital antiplatelet and antithrombotic management. Pre-PCI hospital oral antiplatelet considerations Aspirin Aspirin irreversibly inhibits cyclooxygenase resulting in the inhibition of thromboxane A2 production, thereby reducing the thromboxane A2-mediated amplification of platelet aggregation [11]. Aspirin levels peak within 1 h of oral administration to exert its antiplatelet effect, which highlights the importance of pre PCI hospital administration [12]. Non-enteric coated formulations of aspirin are preferred and patients should be instructed to chew aspirin to increase absorption and bioavailability. As compared to placebo, aspirin monotherapy has been shown to reduce short term vascular mortality after myocardial infarction (MI) (absolute risk reduction of 2.4 %; number needed to treat (NNT) of 47; p \ 0.001) [13]. Antiplatelet therapy administered in the pre-hospital setting improves ST-segment resolution prior to primary PCI and increases rates of arterial patency [14]. The optimal initial dose of aspirin in STEMI remains uncertain. A retrospective study by Berger et al. [15] in 48,422 fibrinolytic-treated STEMI patients showed no difference in 30-day mortality or ischemic-related outcomes in patients treated initially with 162 mg of aspirin as compared with 325 mg of aspirin; however, the higher dose of aspirin was independently associated with greater 123 risk of moderate to severe bleeding (OR 1.14; 95 % CI 1.05–1.24, p = 0.003). There was no difference in 30-day cardiovascular death, MI, or stroke in the Clopidogrel and Aspirin Optimal Dose Usage to Reduce Recurrent Events—Seventh Organization to Assess Strategies in Ischemic Symptoms (CURRENT-OASIS 7) trial that compared open-label high-dose (300–325 mg daily) aspirin to low-dose (75–100 mg) aspirin in patients with acute coronary syndromes undergoing planned PCI [16, 17]. For suspected STEMI, aspirin 162–325 mg as the initial dose should remain the standard, with the exception of patients with true allergy to aspirin [18]. P2Y12 inhibitors These agents work through inhibition of adenosine diphosphate (ADP) induced platelet aggregation by inactivating the P2Y12 receptor on the platelet surface, thereby preventing cross linking, and have led to improved outcomes when combined with aspirin in patients treated with intracoronary stents [19, 20]. Ticlopidine was a first generation ADP receptor antagonist for patients undergoing PCI and as secondary prevention in patients with a history of stroke. However, severe side effects including neutropenia and thrombotic thrombocytopenic purpura combined with twice daily dosing limited its use. Clopidogrel, which is now more commonly used than ticlopidine, is effective in reducing cardiovascular events in high-risk patients with acute coronary syndromes when combined with aspirin. Clopidogrel reduces risk irrespective of upfront PCI or medical therapy, with benefits observed as early as 24 h after initiating therapy [19, 21]. Several clinical trials have subsequently evaluated whether the benefits of clopidogrel treatment in unstable angina/ non-STEMI could also be observed in patients with STEMI. The Clopidogrel and Metoprolol in Myocardial Infarction Trial (COMMIT-CCS 2) randomized [45,000 patients with suspected MI (93 % had STEMI or leftbundle branch block) to clopidogrel 75 mg daily without a loading dose or placebo on a background of antiplatelet therapy (aspirin 162 mg daily) and found a significant reduction in the composite outcome of death, reinfarction, or stroke (OR 0.91; 95 % CI 0.86–0.97; NNT = 112) [22]. Sabatine et al. [23] showed improved ischemic outcomes and greater patency of the infarct-related artery when clopidogrel was added to a standard regimen of fibrinolytic therapy, aspirin, and dose-adjusted unfractionated heparin (UFH) in patients with STEMI. Clopidogrel benefits patients with STEMI receiving fibrinolytic therapy, resulting in a 36 % reduction in the odds of death, recurrent MI, or an occluded infarct artery [24]. In a pre-specified subgroup of the Clopidogrel as Adjunctive Reperfusion Therapy—Thrombolysis in Myocardial Infarction Author's personal copy Pre PCI hospital antithrombotic therapy for ST elevation myocardial infarctions (CLARITY-TIMI 28) trial, 1,863 patients underwent PCI after treatment with fibrinolysis, half of whom received clopidogrel (300 mg loading dose followed by 75 mg daily) while the other half received placebo [25]. Pretreatment with clopidogrel, as compared to placebo, was associated with fewer rates of death, MI, or stroke at 30 days (3.6 vs. 6.2 %; adjusted OR 0.54; 95 % CI 0.35–0.85, NNT = 39) and greater patency in the infarct artery (86.9 vs. 80.8 %, p \ 0.001) [25]. Of note, there is insufficient evidence to support a loading dose of clopidogrel in older adults (age [75 years) undergoing fibrinolysis for STEMI [26]. Although the optimal loading dose for clopidogrel has not clearly been established [27], a 600 mg load as compared to a 300 mg load achieves greater inhibition of platelet aggregation at onset (approximately 2 h) and peak (approximately 6 h), and has been associated with both lower rates of cardiovascular events without an increase in bleeding and improved short term outcomes in STEMI patients undergoing PCI [28, 29]. Clopidogrel, especially when administered with concomitant antiplatelet and antithrombotic therapies, has been associated with increased risk of major bleeding. In the Clopidogrel in Unstable Angina to Prevent Recurrent Events (CURE) trial, the combination of clopidogrel and aspirin as compared to aspirin alone was associated with higher rates of major bleeding (3.7 vs. 2.7 %, respectively; relative risk (RR) 1.38; 95 % CI 1.13–1.67; p = 0.001) and higher rates of blood transfusion C2 U (2.8 vs. 2.2 %, respectively; RR 1.30; 95 % CI 1.04–1.62; p = 0.02), without observed differences in life-threatening or fatal bleeding [21]. Efforts to minimize bleeding with clopidogrel therapy require appropriate dosing of anticoagulants in the acute setting and adhering to lower maintenance doses of aspirin, as stacked antithrombotics markedly increase bleeding risk [30]. Despite large-scale outcome studies supporting the use of clopidogrel across the spectrum of acute coronary syndromes, important limitations of the drug have been identified. Clopidogrel is a prodrug, requiring bioactivation in a 2-step process from several CYP450 enzymes, chiefly involving CYP2C19, to convert the prodrug to its pharmacologically active form (R-130964) [31]. It is important to note that only about 15 % of the prodrug undergoes the process of bioactivation to become potentially available to exert its antiplatelet effect [32]. Observational studies have shown that patients with genetic polymorphisms of CYP2C19, particularly those with two copies of the CYP2C19*2 and CYP2C19*3 loss-of-function alleles, are poor metabolizers of clopidogrel, appear to exhibit less antiplatelet effect after standard clopidogrel doses, and have higher rates of cardiovascular events after MI and PCI [32]. In addition, individual variability in clopidogrel responsiveness may be associated with other genetic and nongenetic factors, including comorbid health conditions, age, body mass, smoking status (via CYP1A2), and medications (via CYP2C9) [33–35]. The association of genetic polymorphisms of CYP2C19 with higher platelet reactivity and higher event rates has prompted interest in genotyping patients, particularly those at higher ischemic risk; however, outcomes data to guide antiplatelet strategy based on results obtained from genotyping are lacking. Finally, identification of CYP2C19 variants still only accounts for a small percentage of the variability in clopidogrel responsiveness [35]. These complex issues have promoted interest in developing more potent ADP receptor blockers with less response variability. Prasugrel, like clopidogrel, is a prodrug and thienopyridine that requires biotransformation to an active metabolite; however, the efficiency of its absorption and metabolic activation are greater than clopidogrel. Conversion to the active metabolite (R-138727) occurs through hydrolysis by intestinal carboxylase, followed by oxidation via the CYP450 system [36]. Importantly, the major CYP subtypes involved in metabolic activation are CYP3A and CYP2B6, with CYP2C19 having less importance, thus obviating the issues related to responsiveness that have been associated with the metabolism of clopidogrel. For prasugrel, this has the net effect of both rapid absorption and metabolism, and attainment of peak plasma concentration of the active metabolite in approximately 30 min after dosing [37]. The Trial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet Inhibition with Prasugrel—Thrombolysis in Myocardial Infarction (TRITON-TIMI 38) compared prasugrel to clopidogrel in moderate-to-high risk acute coronary syndrome patients [38]. In this study, 26 % (n = 3,534) of patients presented with STEMI and the endpoint was the composite outcome of cardiovascular death, MI, or stroke [38]. Prasugrel was administered as a loading dose of 60 mg followed by a maintenance dose of 10 mg daily as compared to clopidogrel at a loading dose of 300 mg followed by 75 mg daily. In the TRITON study, prasugrel, as compared to clopidogrel, was associated with significantly fewer primary outcome events (9.9 vs. 12.1 %; p \ 0.001; NNT = 46) and fewer events of stent thrombosis (1.1 vs. 2.4 %; p \ 0.001; NNT = 77) but at a cost of more major bleeding (2.4 vs. 1.8 %; p = 0.03; NNH = 167), including life-threatening and fatal bleeding. The greater antiplatelet effect with prasugrel increased the susceptibility of bleeding in certain important subgroups such as those age C75 years, prior stroke or transient ischemic attack, or low body weight (\60 kg) such that net clinical benefit with prasugrel as compared to clopidogrel in these patients was not observed. However, these subgroups suffered more bleeding complications irrespective of the treatment assignment [38]. Information regarding pre-hospital 123 Author's personal copy S. M. Gharacholou et al. administration of study medication was not reported in TRITON-TIMI 38, although patients with STEMI and intending to undergo primary PCI could be treated with study medication up to 24 h prior to their procedure. Most patients (74 %) received first dose of study medication after insertion of intracoronary guidewire and up to 1 h after PCI (i.e., over half of patients were loaded after PCI), thus few patients were ‘‘pre-treated’’ with study medication [38, 39]. Importantly, there were no significant differences between treatment assignment with regards to cardiovascular mortality or overall mortality in TRITON-TIMI 38, with results driven by differences in nonfatal MI between treatment groups. Prasugrel should be administered cautiously given the apparent increased bleeding in at-risk subgroups such as the elderly, those with a history of cerebrovascular disease, and low body weight [27, 40]. Ticagrelor is a reversibly binding noncompetitive oral P2Y12 receptor antagonist, does not require metabolic activation, is rapidly absorbed with a maximum plasma concentration at 90 min, and has linear and predictable pharmacokinetics [41]. Ticagrelor was compared to clopidogrel in the Platelet Inhibition and Patient Outcomes (PLATO) trial which tested the hypothesis that ticagrelor would be superior to clopidogrel on the composite outcome of cardiovascular death, MI, or stroke in 18,624 patients with acute coronary syndrome [42]. At 12 months, treatment with ticagrelor (180 mg load followed by 90 mg twice daily), as compared to clopidogrel (300 mg load followed by 75 mg daily), was associated with a lower rate of the composite outcome (9.8 vs. 11.7 %; p \ 0.001; NNT = 53), MI (5.8 vs. 6.9 %; p = 0.005; NNT = 91), and cardiovascular death (4.0 vs. 5.1 %; p = 0.001; NNT = 91). In the subgroup of patients receiving intracoronary stents, treatment with ticagrelor resulted in lower rates of stent thrombosis (1.3 vs. 1.9 %; p = 0.009; NNT = 167). In a secondary analysis of the 41 % of patients from PLATO that presented with STEMI (n = 7,544) and underwent primary PCI, there was a trend towards similar benefits as observed in the parent trial, with a 1.4 % absolute risk reduction (13 % RR reduction) in the primary endpoint favoring ticagrelor (HR 0.87; 95 % CI 0.75–1.01; p = 0.07), including a 0.8 % absolute risk reduction (34 % RR reduction) in definite stent thrombosis with ticagrelor (HR 0.66; 95 % CI 0.45–0.95; p = 0.03) [43]. Adverse events reported in follow up included higher rates of dyspnea in patients treated with ticagrelor that appeared to be self-limiting and non-serious but was associated with a higher rate of study drug discontinuation. There was no difference in major bleeding between patients treated with ticagrelor as compared to patients treated with clopidogrel, including no differences in intracranial hemorrhage, fatal bleeding, or transfusion rates between groups [43]. Non-procedure related major and 123 minor bleeding rates were, however, higher with ticagrelor as compared to clopidogrel (5.1 vs. 3.7 %, respectively; p = 0.02; NNH = 72) [43]. Since the timing from first dose of study drug to PCI in patients with STEMI was a median of 15 min in the PLATO trial, the study was not designed to evaluate the role for administration of ticagrelor in the pre-hospital setting in patients with probable STEMI. Its use in primary PCI are likely to be limited to scenarios where perceived bleeding risk is low, concomitant anticoagulant use has been appropriately dosed and monitored, and the clinician is planning to treat the patient with maintenance doses of aspirin B100 mg daily. If these conditions are satisfied, ticagrelor may become an attractive alternative to clopidogrel due to its superior efficacy and could be adopted in pre-hospital administration of STEMI patients undergoing primary PCI. Glycoprotein IIb/IIIa inhibitors Glycoprotein (GP) IIb/IIIa inhibitors exert their antiplatelet effect through inhibition of the GP IIb/IIIa platelet surface receptor, thus inhibiting the final common pathway in platelet aggregation. The current role for GP IIb/IIIa inhibitors in patients with STEMI referred for primary PCI is less clear, since prior studies of GP IIb/IIIa use in this population did not represent a high percentage that were also being treated on a background of both dual antiplatelet therapy (i.e., aspirin and thienopyridine) and anticoagulation [27]. Therefore, net clinical benefit (benefits minus risks/adverse sequlae) of GP IIb/IIIa inhibitors are less well established in the contemporary era of STEMI patients undergoing primary PCI. Initial development of GP IIb/ IIIa’s in early phase clinical trials were of oral GP IIb/IIIa inhibitors; however, these agents were associated with higher rates of both bleeding and mortality [44]. Abciximab and the small molecule GP IIb/IIIa inhibitors, primarily eptifibatide and tirofiban, have been shown in a pooled meta-analysis of randomized trials and observational registries to have comparable efficacy for reduction of death and MI and similar rates of in-hospital major bleeding in STEMI patients undergoing primary PCI, suggesting non-inferiority of small molecule agents as compared to abciximab [45]. Abciximab, as compared to placebo, did not reduce infarct size or clinical events at 30 days in a contemporary randomized double-blinded study of STEMI patients undergoing PCI who had been pre-treated with 600 mg of clopidogrel and 500 mg of aspirin [46]. The timing of administering GP IIb/IIIa’s in STEMI patients was recently evaluated in 2,453 STEMI patients assigned to half-dose fibrinolysis plus abciximab prior to PCI, abciximab prior to PCI, or abciximab administered at the time of PCI [47]. There were no Author's personal copy Pre PCI hospital antithrombotic therapy for ST elevation myocardial infarctions observed benefits and higher rates of bleeding with upstream abciximab as compared to abciximab at the time of PCI, suggesting that timing for use of GP IIb/IIIa’s should be relegated to selective use in the catheterization lab based on angiographic and clinical factors. Thus, invoking use of GP IIb/IIIa inhibitors at STEMI-referral sites prior to transfer for primary PCI may not improve outcomes and could potentially introduce greater transfer delay due to dosing considerations and use of cumbersome intravenous drips. Administration of fibrinolysis (half or full-dose) upfront with or without GP IIb/IIIa inhibitors with the intent of proceeding directly to PCI has been referred to as facilitated PCI, though current recommendations advise against using labels such as facilitated or rescue in contemporary STEMI management [27]. The use of facilitated PCI in STEMI regional care systems has been described [48]; however, higher rates of bleeding complications remain a significant impediment to more widespread adoption and the totality of current evidence does not support this approach routinely [49]. The addition of GP IIb/IIIa inhibitors to fibrinolysis compared to fibrinolysis alone in a facilitated PCI strategy has been tested [50]. Despite improved infarct-related artery flow at the time of angiography, clinical harm due to ischemic and bleeding complications result in greater risk with a facilitated strategy [49, 50], and current guidelines do not recommend facilitated PCI as a routine reperfusion approach [27]. Instead, the transfer of fibrinolytic treated STEMI patients to the nearest PCI-hospital for possible rescue PCI (i.e., PCI performed after failure of fibrinolysis based on presence of clinical symptoms and incomplete (\50–70 %) resolution of ST-segment elevation) is recommended since at least a 40 % fibrinolytic failure rate can be expected [27]. Pre-hospital anticoagulant considerations There are several anticoagulant options for patients with STEMI referred for PCI, including UFH, enoxaparin, fondaparinux, and bivalirudin (Table 1); however, optimal use of anticoagulation relies on institutional-specific algorithms for managing anticoagulation at the STEMIreceiving center in order to reduce medication and dosing error, optimize anti-ischemic benefit while reducing bleeding risk, and to maintain flexibility in adapting the anticoagulation strategy based on clinically related factors (i.e., renal insufficiency, bleeding risk, adjunctive GP IIb/ IIIa use) [27, 51]. Familiarity with all major anticoagulants is essential given that a proportion of patients co-administered anticoagulants at the time of fibrinolysis may subsequently be referred for rescue PCI. In this case, appropriate anticoagulation at the correct doses will need to be continued during the PCI procedure. The use of UFH for acute MI has demonstrated reductions in mortality and reinfarction in an era prior to standard use of aspirin, yet added benefits for UFH in the era of fibrinolytic therapy have not been convincing [52]. UFH has become the comparator in non-inferiority and activecontrol superiority studies of alternative anticoagulant Table 1 Anticoagulants in the Pre PCI setting in STEMI Anticoagulant Bolus dose Maintenance dose Renal adjustment Cath lab considerations UFH 60 U/kg IV (maximum 4,000 U) 12 U/kg/h (maximum 1,000 U/h) OR may be omitted at non-PCI hospital to minimize transfer delay None Supplemental boluses administered in Cath lab to maintain ACT [ 250 if GP IIb/IIIa use is not planned OR switch to bivalirudin Enoxaparin 30 mg IV (for age \75 years) 1.0 mg/kg SQ Q12H if \75 years old (first dose to be administered 15 min after IV bolus) or 0.75 mg/ kg SQ Q12H if [75 years old Advise against using enoxaparin if SCr [ 2.5 mg/dL in men or [2.0 mg/dL in women If last SQ dose of enoxaparin was given within 8 h, no additional enoxaparin needed. If last SQ dose of enoxaparin was given 8-12 h earlier, administer 0.3 mg/kg IV Fondaparinux 2.5 mg IV (if SCr \ 3.0 mg/ dL) 2.5 mg SQ Q24H Not recommended if SCr [ 3.0 mg/dL (patients were excluded from OASIS-6 [55]) Supplement the fondaparinux-treated patient with anticoagulant that possesses anti-IIa activity (either UFH or bivalirudin). Bivalirudina 0.75 mg/kg IV 1.75 mg/kg/h (infusion usually terminated at end of procedure) Reduced to 1 mg/kg/h (CrCl \ 30 mL/min) or 0.25 mg/kg/h if on dialysis (bolus remains unchanged) Confirm oral antiplatelet therapy (aspirin and thienopyridine) have been administered UFH unfractionated heparin, ACT activated clotting time, GP glycoprotein, SQ subcutaneously, IV intravenously, CrCl creatinine clearance, OASIS Organization for the Assessment of Strategies for Ischemic Syndromes a Primarily started in the cardiac catheterization laboratory upon patient arrival 123 Author's personal copy S. M. Gharacholou et al. strategies, yet drawing conclusions regarding comparative efficacy has been limited due to differences in dosing across studies, characteristics of the study population, differences in duration of therapy in treatment arms, and monitoring strategies of anticoagulant effect [26]. For STEMI, UFH has been a preferred anticoagulant in the pre-hospital setting or when transitioning from the emergency department directly to the catheterization laboratory for primary PCI [18]. The recommended dosing for UFH is 60 U/kg intravenously (not to exceed a 4,000 U bolus) followed by an infusion of 12 U/kg/h (not to exceed 1,000 U/h) with a goal activated partial thromboplastin time of 1.5–2.0 times the local laboratory reference values, which usually approximates 50–70 s [18, 53]; however, the initial bolus may be modified based on whether adjunctive GP IIb/IIIa inhibitors are going to be used. There are limited data for either enoxaparin or fondaparinux as the sole anticoagulant in patients with STEMI undergoing primary PCI, yet these therapies may be started upstream, at the non-PCI hospital, and strategies for switching, supplementing, or continuing the anticoagulant will need to be considered. Enoxaparin, a low molecular weight heparin with a higher ratio of anti-Factor Xa to antiFactor IIa activity than UFH, has been studied in fibrinolytic-treated patients with STEMI, and includes a specific dosing regimen adjusted for age and renal function designed to avoid excess bleeding events due to accumulation of drug and extended anti-Factor Xa activity (Table 1) [54]. For the enoxaparin treated patient with STEMI that is referred for primary PCI and receives the 30 mg IV loading dose prior to transfer, the weight-based supplemental subcutaneous dose should be administered in 15 min by paramedic or emergency department personnel, if not given immediately after the IV loading dose. The rationale for the IV loading dose of enoxaparin in STEMI is related to the pharmacokinetic activity of enoxaparin, which results in peak anti-thrombin and anti-Factor Xa activity 3–5 h after subcutaneous injection. Fondaparinux, a synthetic pentasaccharide that binds antithrombin and inhibits Factor Xa, exerts its antithrombotic effect through antithrombin-mediated neutralization of Factor Xa and subsequent inhibition of thrombin formation. Subcutaneous injection results in near complete bioavailability of the drug and therapeutic drug concentrations are reached approximately 2–3 h post-dose. Fondaparinux has been compared to placebo and UFH in a randomized trial of STEMI patients, including non-reperfused, fibrinolytic-treated, and patients undergoing PCI [55]. In the subset of the stratum that underwent primary PCI, death or reinfarction rates among fondaparinux-treated patients were similar to patients treated with UFH (8.5 vs. 8.2 %, respectively, p = 0.61), yet there were higher rates of guide catheter thrombus events in the fondaparinux 123 group as compared to those treated with UFH (22.0 vs. 0 %, respectively, p \ 0.001) [55]. In addition, there were numerically greater coronary complications in the fondaparinux group as compared to the UFH group (270 vs. 225, respectively, p = 0.04). On this basis, supplemental anticoagulation with UFH or bivalirudin is advised in fondaparinux-treated STEMI patients undergoing planned PCI. If STEMI patients receive fondaparinux at the non-PCI hospital, it is critical to communicate this information to the PCI hospital so that appropriate supplemental antithrombotic therapy with anti-IIa activity can be provided to support the PCI procedure (Table 1). Bivalirudin has recently emerged as an important anticoagulant in primary PCI for STEMI. In the Harmonizing Outcomes with Revascularization and Stents in Acute Myocardial Infarction (HORIZONS-AMI) trial, bivalirudin (with provisional GP IIb/IIIa inhibitor for no-reflow or thrombus after PCI) was compared to UFH plus a GP IIb/ IIIa inhibitor (dose-adjusted eptifibatide or abciximab per the investigator’s discretion) on rates of major bleeding or major adverse cardiovascular events at 30-days [56]. Bivalirudin resulted in a lower rate of the primary endpoint (9.2 vs. 12.1 %, p = 0.005), driven by less major bleeding, and an observed lower rate of cardiac death and all-cause mortality at 30-days. There was a higher rate of acute (\24 h) stent thrombosis in the bivalirudin treated patients as compared to the UFH/GP IIb/IIIa group (1.3 vs. 0.3 %, p \ 0.001). Some of the higher rate of stent thrombosis may have been explained by a lower loading dose of clopidogrel (300 mg instead of 600 mg) and bivalirudin monotherapy [27]. Thus, from a pre-hospital STEMI activation standpoint in patients being referred for primary PCI, it is important to recognize that the majority of patients in HORIZONS-AMI (76 %) were treated with open label UFH as the initial anticoagulant, with subsequent secondary analyses suggesting that switching from UFH to bivalirudin, as compared to continued use of UFH with a GP IIb/IIIa inhibitor, may not only be associated with lower rates of major bleeding, but also lower longterm cardiac mortality and reinfarction [57]. Indeed, in the interventional setting, patients initially treated with UFH can be readily converted to bivalirudin to support the primary PCI procedure, with a proviso of a 30 min interval between dosing of the anticoagulants [56]. Antithrombotic approaches adopted by regional primary PCI programs Early primary PCI for STEMI and its time-dependant benefits have been known for some time [58]. Implementing primary PCI in the US requires collaboration and direct communication between EMSs, emergency medicine physicians, and cardiologists. Additional complexities of Author's personal copy Pre PCI hospital antithrombotic therapy for ST elevation myocardial infarctions delivering primary PCI in the US include resource utilization, staffing personnel, and economic and logistical details regarding cardiac services in rural hospitals [6, 7]. Several organizations have developed standardized protocols within their regional network for antithrombotic therapies to be administered before or at the time of primary PCI that aims for efficiency, simplicity, and an evidence based approach to STEMI care (Table 2). These programs have been effective in integrating rural community hospitals with a PCI center [48, 59–62], and have been implemented statewide [59] and across state borders [60]. One important limitation in STEMI care in the US has been a lack of standardized guideline-driven protocols. Instead, protocols tend to vary from region to region. Variable dosing of antithrombotics, selection of thienopyridines versus GP IIb/IIIa inhibitors for adjunctive antiplatelet therapy, and select use of facilitated PCI are examples that make outcome comparisons across regional protocols difficult (Table 2). Notably, achieving goals and benchmarks on pre-PCI hospital care and transport appears feasible in the conduct of clinical trials [63]; however, achieving these goals and benchmarks in the ‘‘real-world’’ setting has proven to be problematic. Registry studies show that \9 % of patients have total (e.g., arrival at non-PCI hospital to first device at PCI hospital) door-to-balloon times of \90 min [64]. It would appear that the same level of coordination and motivation that drives multi-center clinical trials is what is required to institute protocol standards for STEMI care in the patient care setting. Streamlining, simplifying, and maintaining efficacy Specific factors that are unique to certain regions must be taken into account when designing standardized STEMI protocols. For example, inclement weather directly impacts the selection of transport strategies, which may be more of a factor in the upper Midwest than in the Southeastern US during the winter months. The local distribution of EMS services including staffed helicopter units, paramedic units, or ‘‘basic’’ EMS units can affect time to STEMI recognition on the electrocardiogram and qualifications to administer antithrombotic agents, for example. Variations exist in level of integration between health providers and facilities, governance of local emergency systems, and willingness to invest in regional STEMI system of care. However, efforts to understand strategies that work and to allow for comparative evaluation between programs Table 2 Antithrombotic recommendations suggested for use by PCI centers for interhospital transfer patients with STEMI intended for primary PCI Program name Program location Reperfusion of Acute Myocardial Infarction in North Carolina Emergency Departments (RACE) [59] State of NC Geisenger STEMI protocol [62, 65] Stat Heart Program [61] Minneapolis Heart Institute Level 1 Myocardial Infarction Program [48] Mayo Clinic STEMI Protocol [60] Antiplatelet strategy Anticoagulant strategy Aspirin 162 mg to 325 mg Bolus: UFH 70 U/kg Clopidogrel 300 mg to 600 mg or Prasugrel 60 mga Maintenance: None during transfer Central PA Aspirin 325 mg Clopidogrel 600 mg Bolus: UFH 70 U/kg Maintenance: None during transfer Central IL Aspirin 324 mg Clopidogrel 300 mg Bolus: UFH 70 U/kg (maximum of 7,000 U) GP IIb/IIIa inhibitor (abciximab or eptifibatide) Maintenance: 15 U/kg/h (maximum of 1,000 U/h) Regional sites within 200 miles of Minneapolis, MN Aspirin 325 mg Bolus: UFH 60 U/kg (maximum of 4,000 U) Regional sites within 150 miles of Rochester, MN Aspirin regimen not reported but high rates of use ([95 %) Clopidogrel 600 mg Maintenance: 12 U/kg/h (maximum of 1,000 U/h) Clopidogrel not used University of Wisconsin-Madison Regional sites within 100 miles of Madison, WI Aspirin 325 mg Clopidogrel 600 mg Bolus: UFH 60 U/kg (maximum of 4,000 U) Maintenance: 12 U/kg/h (maximum of 1,000 U/h) Bolus: UFH 60 U/kg (maximum of 4,000 U) Maintenance: None during transfer UFH unfractionated heparin, GP glycoprotein a Operations manual (available at http://www.nccacc.org) 123 Author's personal copy S. M. Gharacholou et al. require consensus on antithrombotic strategies for STEMI patients referred for primary PCI. At the University of Wisconsin-Madison, paramedics in Dane County (Madison, WI) activate the cardiac catheterization laboratory based on their own interpretation of electrocardiogram acquired at the scene, and administer a standard pre-treatment regimen of antithrombotic therapy en route (Table 2). Adjunctive medications, such as nitroglycerin and analgesics, are also administered for symptom relief. For STEMI outside of Dane County, the decision to travel directly by ground or helicopter (Med Flight) from the regional non-PCI hospital, or through a Med Flight intercept (i.e., convenient landing zone near the scene) is rapidly determined by radio communication between emergency room physicians and Med Flight dispatch. Factors such as flight conditions, road conditions, and travel distances are rapidly assessed. The University of Wisconsin-Madison Med Flight Helicopter transport program is uniquely staffed by board-certified emergency physicians (MD/DO) that significantly enhances the team’s Fig. 1 STEMI referral sites and their geographic relationship to the University of WisconsinMadison as represented by ground distance (mileage) and nautical miles (rings) 123 capability to make clinical decisions and institute care related to pre PCI hospital antithrombotics. The geographic distribution of non-PCI centers in our STEMI referral program at the University of Wisconsin-Madison is illustrated in Fig. 1. In our program, we initiate antiplatelet therapy with aspirin 325 mg chewed at first contact with EMS. In patients unable to take oral agents and who do not have a nasogastric tube in place, a 300 mg aspirin suppository can be administered [9]. For fibrinolytic treated patients in the non-PCI emergency room, a clopidogrel loading dose of 300 mg in patients B75 years of age is administered. A strategy of administering clopidogrel 75 mg without a loading dose can be used in patients [75 years old with STEMI undergoing fibrinolysis, but careful attention to the adjunctive anticoagulant dose is imperative [22]. For primary PCI, we recommended rapid clopidogrel loading of 600 mg, as has been used in contemporary primary PCI trials [46]. However, it should be noted that the optimal clopidogrel loading dose in patients with STEMI referred for primary PCI has Author's personal copy Pre PCI hospital antithrombotic therapy for ST elevation myocardial infarctions not been conclusively determined from clinical trials [27]. Our choice of clopidogrel is based on the favorable safety profile (in comparison to prasugrel or ticagrelor), regional provider familiarity, and regional ease of access to the drug. On arrival in the STEMI receiving hospital’s cardiac catheterization lab, a formal checklist is entered that includes queries that aspirin and clopidogrel were administered, their doses and time of administration. This approach is critical to avoid the untenable scenario of stacked dosing that may increase bleeding risk or omission that may lead to recurrent ischemic events or stent thrombosis [18]. Our recommended anticoagulant has remained a bolus of UFH without maintenance infusion, similar to the anticoagulant of choice among other regional care systems, and dose of anticoagulant is verified as part of our checklist upon patient arrival to the cath lab. However, slight differences in dosing regimens may be due to patient-related factors such as obesity or perceived bleeding risk, or whether some programs prefer starting maintenance infusions post bolus (Table 2). We routinely switch to bivalirudin during the PCI procedure. A greater percentage of our procedures are being performed via the transradial approach, which in the future may enable intensifying the antithrombotic regimen or even a re-evaluation of facilitated PCI in selected patients. Of important note, the above-described regional STEMI protocol has been agreed upon by the three STEMI-receiving PCI hospitals in Madison, WI. Such integration is critical for regional providers to avoid problems associated with conflicting protocols. However, a standardized protocol does not usurp clinical experience or sound clinical judgment for selecting appropriate adjunctive pharmacological therapies or refining dosing regimens based on a risk–benefit calculus for individual patients. Summary Standardized, system wide approaches to STEMI care involve the creation of regional networks, and multi-disciplinary collaborations to improve STEMI outcomes. Time-dependant benefits of antithrombotic agents instituted in the patient with STEMI are important co-interventions in the delivery of reperfusion therapy. Goals should include reducing delay to coronary reperfusion, achieving PCI as the preferred reperfusion strategy within 90 min of first medical contact, and preventing medication dosing errors, and improving quality and performance. Acknowledgments We would like to thank Vicki Carter and Renae Buchheim for their assistance in creating the figure of the regional map of STEMI referral centers that coordinate care with the University of Wisconsin-Madison. No compensation, apart from employment at the University of Wisconsin-Madison Hospitals and Clinics, was provided. References 1. Antman EM, Anbe DT, Armstrong PW et al (2004) ACC/AHA guidelines for the management of patients with ST-elevation myocardial infarction: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Committee to Revise the 1999 Guidelines for the Management of Patients with Acute Myocardial Infarction). Circulation 110:e82–292 2. Jacobs AK, Antman EM, Faxon DP, Gregory T, Solis P (2007) Development of systems of care for ST-elevation myocardial infarction patients: executive summary. Circulation 116:217–230 3. Gharacholou SM, Alexander KP, Chen AY et al (2010) Implications and reasons for the lack of use of reperfusion therapy in patients with ST-segment elevation myocardial infarction: findings from the CRUSADE initiative. Am Heart J 159:757–763 4. Eagle KA, Goodman SG, Avezum A, Budaj A, Sullivan CM, Lopez-Sendon J (2002) Practice variation and missed opportunities for reperfusion in ST-segment-elevation myocardial infarction: findings from the Global Registry of Acute Coronary Events (GRACE). Lancet 359:373–377 5. Grzybowski M, Clements EA, Parsons L et al (2003) Mortality benefit of immediate revascularization of acute ST-segment elevation myocardial infarction in patients with contraindications to thrombolytic therapy: a propensity analysis. JAMA 290:1891– 1898 6. Boden WE, Eagle K, Granger CB (2007) Reperfusion strategies in acute ST-segment elevation myocardial infarction: a comprehensive review of contemporary management options. J Am Coll Cardiol 50:917–929 7. Jacobs AK, Antman EM, Ellrodt G et al (2006) Recommendation to develop strategies to increase the number of ST-segment-elevation myocardial infarction patients with timely access to primary percutaneous coronary intervention. Circulation 113:2152–2163 8. Goodman SG, Menon V, Cannon CP, Steg G, Ohman EM, Harrington RA (2008) Acute ST-segment elevation myocardial infarction: American College of Chest Physicians EvidenceBased Clinical Practice Guidelines (8th Edition). Chest 133: 708S–775S 9. Van de Werf F, Bax J, Betriu A et al (2008) Management of acute myocardial infarction in patients presenting with persistent STsegment elevation: the Task Force on the Management of STSegment Elevation Acute Myocardial Infarction of the European Society of Cardiology. Eur Heart J 29:2909–2945 10. Cantor WJ, Fitchett D, Borgundvaag B et al (2009) Routine early angioplasty after fibrinolysis for acute myocardial infarction. N Engl J Med 360:2705–2718 11. Patrono C (1994) Aspirin as an antiplatelet drug. N Engl J Med 330:1287–1294 12. Eisenberg MJ, Topal EJ (1996) Prehospital administration of aspirin in patients with unstable angina and acute myocardial infarction. Arch Intern Med 156:1506–1510 13. ISIS-2 (Second International Study of Infarct Survival) Collaborative Group (1988) Randomised trial of intravenous streptokinase, oral aspirin, both, or neither among 17,187 cases of suspected acute myocardial infarction: ISIS-2. Lancet 2:349–360 14. Heestermans T, van ’t Hof AW, ten Berg JM et al (2010) The golden hour of prehospital reperfusion with triple antiplatelet therapy: a sub-analysis from the Ongoing Tirofiban in Myocardial Evaluation 2 (On-TIME 2) trial early initiation of triple antiplatelet therapy. Am Heart J 160:1079–1084 15. Berger JS, Stebbins A, Granger CB et al (2008) Initial aspirin dose and outcome among ST-elevation myocardial infarction patients treated with fibrinolytic therapy. Circulation 117: 192–199 123 Author's personal copy S. M. Gharacholou et al. 16. Mehta SR, Bassand JP, Chrolavicius S et al (2010) Dose comparisons of clopidogrel and aspirin in acute coronary syndromes. N Engl J Med 363:930–942 17. Mehta SR, Tanguay JF, Eikelboom JW et al (2010) Double-dose versus standard-dose clopidogrel and high-dose versus low-dose aspirin in individuals undergoing percutaneous coronary intervention for acute coronary syndromes (CURRENT-OASIS 7): a randomised factorial trial. Lancet 376:1233–1243 18. Pollack CV Jr, Antman EM, Hollander JE (2008) 2007 focused update to the ACC/AHA guidelines for the management of patients with ST-segment elevation myocardial infarction: implications for emergency department practice. Ann Emerg Med 52(344–355):e341 19. Mehta SR, Yusuf S, Peters RJ et al (2001) Effects of pretreatment with clopidogrel and aspirin followed by long-term therapy in patients undergoing percutaneous coronary intervention: the PCICURE study. Lancet 358:527–533 20. Leon MB, Baim DS, Popma JJ et al (1998) A clinical trial comparing three antithrombotic-drug regimens after coronaryartery stenting. Stent Anticoagulation Restenosis Study Investigators. N Engl J Med 339:1665–1671 21. Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni G, Fox KK (2001) Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation. N Engl J Med 345:494–502 22. Chen ZM, Jiang LX, Chen YP et al (2005) Addition of clopidogrel to aspirin in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet 366:1607–1621 23. Sabatine MS, Cannon CP, Gibson CM et al (2005) Addition of clopidogrel to aspirin and fibrinolytic therapy for myocardial infarction with ST-segment elevation. N Engl J Med 352: 1179–1189 24. Scirica BM, Sabatine MS, Morrow DA et al (2006) The role of clopidogrel in early and sustained arterial patency after fibrinolysis for ST-segment elevation myocardial infarction: the ECG CLARITY-TIMI 28 Study. J Am Coll Cardiol 48:37–42 25. Sabatine MS, Cannon CP, Gibson CM et al (2005) Effect of clopidogrel pretreatment before percutaneous coronary intervention in patients with ST-elevation myocardial infarction treated with fibrinolytics: the PCI-CLARITY study. JAMA 294:1224–1232 26. Antman EM, Hand M, Armstrong PW et al (2008) 2007 Focused Update of the ACC/AHA 2004 Guidelines for the Management of Patients With ST-Elevation Myocardial Infarction: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines: developed in collaboration With the Canadian Cardiovascular Society endorsed by the American Academy of Family Physicians: 2007 Writing Group to Review New Evidence and Update the ACC/AHA 2004 Guidelines for the Management of Patients With ST-Elevation Myocardial Infarction, Writing on Behalf of the 2004 Writing Committee. Circulation 117:296–329 27. Kushner FG, Hand M, Smith SC Jr et al (2009) 2009 focused updates: ACC/AHA guidelines for the management of patients with ST-elevation myocardial infarction (updating the 2004 guideline and 2007 focused update) and ACC/AHA/SCAI guidelines on percutaneous coronary intervention (updating the 2005 guideline and 2007 focused update) a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol 54:2205–2241 28. Levine GN, Bates ER, Blankenship JC et al (2011) ACCF/AHA/ SCAI Guideline for Percutaneous Coronary Intervention. A report of the American College of Cardiology Foundation/ American Heart Association Task Force on Practice Guidelines and the Society for Cardiovascular Angiography and Interventions. J Am Coll Cardiol 58:e44–122 123 29. Mangiacapra F, Muller O, Ntalianis A et al (2010) Comparison of 600 versus 300-mg clopidogrel loading dose in patients with STsegment elevation myocardial infarction undergoing primary coronary angioplasty. Am J Cardiol 106:1208–1211 30. Alexander KP, Chen AY, Roe MT et al (2005) Excess dosing of antiplatelet and antithrombin agents in the treatment of non-ST-segment elevation acute coronary syndromes. JAMA 294:3108–3116 31. Mega JL, Close SL, Wiviott SD et al (2009) Cytochrome P450 genetic polymorphisms and the response to prasugrel: relationship to pharmacokinetic, pharmacodynamic, and clinical outcomes. Circulation 119:2553–2560 32. Holmes DR Jr, Dehmer GJ, Kaul S, Leifer D, O’Gara PT, Stein CM (2010) ACCF/AHA Clopidogrel clinical alert: approaches to the FDA ‘‘boxed warning’’: a report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus Documents and the American Heart Association. Circulation 122:537–557 33. Desai NR, Mega JL, Jiang S, Cannon CP, Sabatine MS (2009) Interaction between cigarette smoking and clinical benefit of clopidogrel. J Am Coll Cardiol 53:1273–1278 34. Gilard M, Arnaud B, Cornily JC et al (2008) Influence of omeprazole on the antiplatelet action of clopidogrel associated with aspirin: the randomized, double-blind OCLA (Omeprazole CLopidogrel Aspirin) study. J Am Coll Cardiol 51:256–260 35. Shuldiner AR, O’Connell JR, Bliden KP et al (2009) Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy. JAMA 302:849–857 36. Small DS, Farid NA, Payne CD et al (2010) Effect of intrinsic and extrinsic factors on the clinical pharmacokinetics and pharmacodynamics of prasugrel. Clin Pharmacokinet 49:777–798 37. Farid NA, Smith RL, Gillespie TA et al (2007) The disposition of prasugrel, a novel thienopyridine, in humans. Drug Metab Dispos 35:1096–1104 38. Wiviott SD, Braunwald E, McCabe CH et al (2007) Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 357:2001–2015 39. Serebruany VL (2011) Timing of thienopyridine loading and outcomes in the TRITON trial: the FDA Prasugrel Action Package outlook. Cardiovasc Revasc Med 12:94–98 40. Bhatt DL (2007) Intensifying platelet inhibition—navigating between Scylla and Charybdis. N Engl J Med 357:2078–2081 41. Teng R, Oliver S, Hayes MA, Butler K (2010) Absorption, distribution, metabolism, and excretion of ticagrelor in healthy subjects. Drug Metab Dispos 38:1514–1521 42. Wallentin L, Becker RC, Budaj A et al (2009) Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 361:1045–1057 43. Steg PG, James S, Harrington RA et al (2010) Ticagrelor versus clopidogrel in patients with ST-elevation acute coronary syndromes intended for reperfusion with primary percutaneous coronary intervention: A Platelet Inhibition and Patient Outcomes (PLATO) trial subgroup analysis. Circulation 122:2131–2141 44. Cannon CP (2000) Learning from the recently completed oral glycoprotein IIb/IIIa receptor antagonist trials. Clin Cardiol 23(Suppl 6):VI-14–VI-17 45. Ottani F, La Vecchia L, De Vita M, Catapano O, Tarantino F, Galvani M (2010) Comparison by meta-analysis of eptifibatide and tirofiban to abciximab in patients with ST-elevation myocardial infarction treated with primary percutaneous coronary intervention. Am J Cardiol 106(167–174):e161 46. Mehilli J, Kastrati A, Schulz S et al (2009) Abciximab in patients with acute ST-segment-elevation myocardial infarction undergoing primary percutaneous coronary intervention after clopidogrel loading: a randomized double-blind trial. Circulation 119:1933–1940 Author's personal copy Pre PCI hospital antithrombotic therapy for ST elevation myocardial infarctions 47. Ellis SG, Tendera M, de Belder MA et al (2008) Facilitated PCI in patients with ST-elevation myocardial infarction. N Engl J Med 358:2205–2217 48. Henry TD, Sharkey SW, Burke MN et al (2007) A regional system to provide timely access to percutaneous coronary intervention for ST-elevation myocardial infarction. Circulation 116:721–728 49. Keeley EC, Boura JA, Grines CL (2006) Comparison of primary and facilitated percutaneous coronary interventions for ST-elevation myocardial infarction: quantitative review of randomised trials. Lancet 367:579–588 50. Investigtors A-P (2006) Primary versus tenecteplase-facilitated percutaneous coronary intervention in patients with ST-segment elevation acute myocardial infarction (ASSENT-4 PCI): randomised trial. Lancet 367:569–578 51. Gharacholou SM, Lopes RD, Washam JB, Newby LK, James SK, Alexander JH (2010) Antithrombotic therapy in acute coronary syndromes: guidelines translated for the clinician. J Thromb Thrombolysis 29:516–528 52. Mahaffey KW, Granger CB, Collins R et al (1996) Overview of randomized trials of intravenous heparin in patients with acute myocardial infarction treated with thrombolytic therapy. Am J Cardiol 77:551–556 53. Antman EM, Hand M, Armstrong PW et al (2008) 2007 focused update of the ACC/AHA 2004 guidelines for the management of patients with ST-elevation myocardial infarction: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol 51:210–247 54. Antman EM, Morrow DA, McCabe CH et al (2006) Enoxaparin versus unfractionated heparin with fibrinolysis for ST-elevation myocardial infarction. N Engl J Med 354:1477–1488 55. Yusuf S, Mehta SR, Chrolavicius S et al (2006) Effects of fondaparinux on mortality and reinfarction in patients with acute ST-segment elevation myocardial infarction: the OASIS-6 randomized trial. JAMA 295:1519–1530 56. Stone GW, Witzenbichler B, Guagliumi G et al (2008) Bivalirudin during primary PCI in acute myocardial infarction. N Engl J Med 358:2218–2230 57. Dangas GD, Mehran R, Nikolsky E et al (2011) Effect of switching antithrombin agents for primary angioplasty in acute myocardial infarction: the HORIZONS-SWITCH analysis. J Am Coll Cardiol 57:2309–2316 58. Berger PB, Ellis SG, Holmes DR Jr et al (1999) Relationship between delay in performing direct coronary angioplasty and early clinical outcome in patients with acute myocardial infarction: results from the global use of strategies to open occluded arteries in Acute Coronary Syndromes (GUSTO-IIb) trial. Circulation 100:14–20 59. Jollis JG, Roettig ML, Aluko AO et al (2007) Implementation of a statewide system for coronary reperfusion for ST-segment elevation myocardial infarction. JAMA 298:2371–2380 60. Ting HH, Rihal CS, Gersh BJ et al (2007) Regional systems of care to optimize timeliness of reperfusion therapy for ST-elevation myocardial infarction: the Mayo Clinic STEMI Protocol. Circulation 116:729–736 61. Aguirre FV, Varghese JJ, Kelley MP et al (2008) Rural interhospital transfer of ST-elevation myocardial infarction patients for percutaneous coronary revascularization: the Stat Heart Program. Circulation 117:1145–1152 62. Blankenship JC, Haldis TA, Wood GC, Skelding KA, Scott T, Menapace FJ (2007) Rapid triage and transport of patients with ST-elevation myocardial infarction for percutaneous coronary intervention in a rural health system. Am J Cardiol 100:944–948 63. Andersen HR, Nielsen TT, Rasmussen K et al (2003) A comparison of coronary angioplasty with fibrinolytic therapy in acute myocardial infarction. N Engl J Med 349:733–742 64. Chakrabarti A, Krumholz HM, Wang Y, Rumsfeld JS, Nallamothu BK (2008) Time-to-reperfusion in patients undergoing interhospital transfer for primary percutaneous coronary intervention in the U.S: an analysis of 2005 and 2006 data from the National Cardiovascular Data Registry. J Am Coll Cardiol 51:2442–2443 65. Blankenship JC, Scott TD, Skelding KA et al (2011) Door-toballoon times under 90 min can be routinely achieved for patients transferred for ST-segment elevation myocardial infarction percutaneous coronary intervention in a rural setting. J Am Coll Cardiol 57:272–279 123