Skip to main content

Advertisement

ADVERTISEMENT

Contemporary View of the Acute Coronary Syndromes (Part I of II)

Ali Moustapha, MD and *H. Vernon Anderson, MD
February 2003
Acute coronary syndromes (ACS) represent a clinical spectrum that extends all the way from unstable angina presenting with worsening episodes of chest pain, to non-ST segment elevation myocardial infarction (NSTEMI) with more prolonged chest pain and biochemical evidence of myocardial injury, to ST-segment elevation myocardial infarction (STEMI) with more extensive myocardial damage and usually the formation of Q-waves on the surface electrocardiogram, and finally to sudden cardiac death. Pathophysiologic correlations in ACS include minor plaque ulceration and transient thrombus formation in unstable angina, more extensive thrombosis in non-Q wave MI, and complete occlusion in STEMI and sudden death. In this review, we will limit the discussion to unstable angina and NSTEMI, and describe some recent developments and new insights into the basic mechanisms thought to underlie ACS. We will address the contemporary antithrombotic approach to treatment of this disorder, including the use of aspirin, thienopyridine agents, unfractionated and low-molecular-weight heparins, and glycoprotein (GP) IIb/IIIa receptor antagonists. In addition, we will address new therapeutic options that may target the inflammatory component of ACS (Table 1). The dilemma about the optimal approach for the treatment of ACS, whether invasive or conservative, is also briefly discussed. Pathophysiology of ACS. Atherosclerotic plaque rupture with thrombus formation is the primary pathophysiologic mechanism behind ACS.1–5 This process is complex. The thrombotic portion starts with exposure of subendothelial constituents like collagen, von Willebrand factor, fibronectin and vitronectin. These elements are recognized by platelet surface receptor GP Ib and lead to platelet activation and adhesion to the vessel wall. Activated platelets release from their alpha granules substances such as thromboxane A2, thrombin, adenosine diphosphate and serotonin, which lead to further platelet activation and aggregation. The final common pathway of platelet aggregation involves activation of the GP IIb/IIIa surface receptors, which cross-link platelets via fibrinogen bridges. Aggregated platelets facilitate the conversion of prothrombin to thrombin; this leads to increased thrombin formation, which in turn is a potent agonist for further platelet activation. Finally, the thrombus is stabilized by thrombin-mediated conversion of fibrinogen to fibrin. Inflammation and factors leading to plaque rupture. Recent studies have shown that inflammatory response to endothelial injury plays an important and possibly decisive role in the pathophysiology and vulnerability of the atherosclerotic plaque. The presence and degree of inflammation and the shift to a procoagulant state, defined by elevated C-reactive protein (CRP), fibrinogen, interleukin (IL)-1 and IL-6, and plasminogen activator inhibitor (PAI), have been strongly associated with an increased risk of future events.6–10 Histologic analysis shows that ruptured plaques exhibit an abundance of inflammatory cells, indicating a causal link. T-lymphocytes, for example, secrete gamma interferon, a cytokine that reduces collagen synthesis by smooth muscle cells.11 Macrophages, on the other hand, secrete matrix metalloproteinases, enzymes responsible for collagen degradation.12 Both T-lymphocytes and macrophages thus contribute to weakening of the collagen matrix and engender subsequent plaque rupture. Further indirect support of the role of inflammation emanates from studies showing that detecting heat released by inflammatory cells within an atherosclerotic plaque predicts future instability and rupture.13 In addition to a weak matrix and presence of inflammatory cells, other studies show that as the size of the soft lipid core increases and the fibrous cap decreases, circumferential stress will increase, particularly in the shoulder region of an atheroma, thus leading to its rupture.14 Aspirin. Aspirin exerts its antiplatelet effects by inhibition of the synthesis of thromboxane A2, a potent amplifier of platelet aggregatory response. At the pharmacological level, aspirin irreversibly acetylates and inactivates prostaglandin G/H synthase, therefore preventing the conversion of arachidonic acid to thromboxane A2. This effect is permanent for the life of the anucleate platelet. Aspirin therapy decreases the incidence of adverse cardiovascular events in patients with stable coronary artery disease, acute STEMI, and in the setting of percutaneous coronary angioplasty. In patients with ACS, four randomized trials have shown that aspirin therapy, in doses ranging from 75–1,320 mg daily, reduces the incidence of death or myocardial infarction by 30–50%.15–18 Aspirin therapy has its limitations. Platelet aggregation caused by adenosine, serotonin, catecholamines and shear stress is not completely inhibited by aspirin administration. More importantly, recent studies suggest that up to 8–12% of patients might be resistant to the antiplatelet effect of aspirin therapy,19 possibly because of its inability to completely suppress thromboxane A2 synthesis.20 Bedside platelet function tests may assist in identifying these patients, although more research is needed. Clopidogrel. Clopidogrel is a thienopyridine agent that blocks adenosine diphosphate-induced platelet aggregation and is analogous to a formerly used drug, ticlopidine. Compared to ticlopidine, however, clopidogrel has a faster onset and longer duration of action, and is associated with a better side-effect profile. In the CAPRIE trial,21 over 19,000 patients with coronary, peripheral or cerebral vascular disease were randomized to aspirin (325 mg/day) or clopidogrel (75 mg/day). Clopidogrel was shown to be modestly superior in the prevention of adverse cardiovascular events and ischemic stroke compared to aspirin (5.83% versus 5.32%; p = 0.043). More importantly, clopidogrel was not associated with an increased incidence of bone marrow suppression on gastrointestinal intolerance compared to aspirin. The CLASSICS trial,22 on the other hand, established that clopidogrel is equivalent to ticlopidine in patients who undergo stenting procedures, but with much better tolerability. The CURE trial23,24 was a randomized, double-blind trial in which 12,562 patients with ACS were randomized within 24 hours after the onset of symptoms to receive either a bolus dose of clopidogrel (300 mg) followed by 75 mg per day for 3–12 months, or matching placebo. Both groups received aspirin. The primary outcome of death from cardiovascular causes, nonfatal myocardial infarction or stroke occurred in 9.3% of the patients in the clopidogrel group and 11.4% of the patients in the placebo group (p Unfractionated heparin. Unfractionated heparin exerts its antithrombotic effects by binding to antithrombin III and inhibiting thrombin action. Data regarding the use of unfractionated heparin in patients with ACS have been conflicting. Several studies have shown that treatment with unfractionated heparin in addition to aspirin produces a trend toward a reduction in the risk of death or non fatal myocardial infarction.17,18,26,27 On the other hand, in a study by Holdright et al.,28 unfractionated heparin did not have any additional benefit over aspirin alone. In that study, however, patients received unfractionated heparin for only 2 days. In a different study by Gurfinkel et al.,29 aspirin alone or aspirin combined with unfractionated heparin were similar and both were inferior to a combination of aspirin and low-molecular-weight heparin. A recent meta-analysis30 showed that the combination of aspirin and unfractionated heparin is associated with a 33% reduction of the incidence of death and non fatal myocardial infarction compared to aspirin alone. This reduction, although not statistically significant, provides some support for the use of this combination in ACS. Low-molecular-weight heparin. Unlike unfractionated heparin, low-molecular-weight heparins exert their anticoagulant action mainly directed against factor Xa, and thus may limit rebound phenomenon by preventing thrombin regeneration after drug discontinuation. Other advantages of low-molecular-weight heparins include better bioavailability, longer duration of action, less binding to plasma proteins and endothelial cells, and resistance to inactivation by PF4. Disadvantages of low-molecular-weight heparins include lack of effective monitoring, especially in patients in the catheterization laboratory, as well as higher costs. Three low-molecular-weight heparins have been evaluated in 7 clinical trials in patients with ACS: enoxaparin,31–33 dalteparin34–37 and nadroparin.29,38 Only one low-molecular-weight heparin, enoxaparin, has been shown to be superior to unfractionated heparin in the medical phase of treatment of ACS. Both the ESSENCE trial31 and the TIMI-11B trial32 randomly assigned patients with angina at rest or non-Q wave myocardial infarction to receive either enoxaparin or continuous intravenous unfractionated heparin. The primary endpoint (death, myocardial infarction or urgent revascularization) occurred less in the enoxaparin group in both studies. A prospectively planned meta-analysis33 of the above two trials showed that enoxaparin was associated with a 20% reduction in death and serious cardiac ischemic events that appeared within the first few days of treatment, and this benefit was sustained through 43 days (Figure 2). Enoxaparin’s treatment benefit was not associated with an increase in major hemorrhage during the acute phase of therapy, but there was a slight increase in the rate of minor hemorrhage. Other low-molecular-weight heparins have also been studied, but results have not been as good as enoxaparin. Based on the FRIC34 FRISC,35 FRISC II,36,37 and FRAXIS38 trials, as well as a study by Gurfinkel at al.,29 both dalteparin and nadroparin seem to be as effective and safe as unfractionated heparin. Possible explanations for the divergent results seen among these three agents include different clinical activities among agents, different trial objectives, designs and endpoints, different patient selection criteria, and different regimens and duration of low-molecular-weight heparins. Platelet glycoprotein IIb/IIIa therapy. The GP IIb/IIIa receptor on the platelet surface membrane constitutes the final common pathway of platelet aggregation. This receptor binds circulating fibrinogen or von Willebrand factor and cross-links adjacent platelets together. Three different GP IIb/IIIa receptor antagonists are currently approved for clinical use. Abciximab is a monoclonal antibody fragment with a high binding affinity for the GP IIb/IIIa receptor, but with additional activity against the vitronectin and MAC-1 receptors. Eptifibatide and tirofiban are small-molecule agents with exclusive specificity for the GP IIb/IIIa receptor. Unlike abciximab, these agents have less binding affinity and a shorter biological half-life. Several trials have established the beneficial effects of GP IIb/IIIa receptor antagonists in patients undergoing percutaneous interventions. In patients with ACS, however, trials of GP IIb/IIIa receptor antagonists have yielded only modest benefits, and sometimes even conflicting results. These include the PRISM,39 PRISM-PLUS,40 PURSUIT,41 PARAGON,42 and GUSTO-IV43 trials (Table 2). A recent meta-analysis44 estimated that these agents reduce the composite endpoint of death or myocardial infarction at 30 days by only 12%, with a marked heterogeneity between different trials (Figure 3). Several analyses have identified certain subgroups of high-risk patients with ACS who would benefit most from GP IIb/IIIa receptor antagonist therapy. For example, when a percutaneous intervention is performed, the benefit of this therapy is more pronounced. This is shown by the 31%, 35% and 42% reductions in the 30-day incidence of death or myocardial infarction seen when a percutaneous intervention was performed in the PURSUIT,45 PARAGON-B, and PRISM-PLUS trials, respectively, as compared to only 6%, 7% and 12% reductions when a percutaneous intervention was not performed. These results are confirmed in the meta-analysis mentioned earlier,44 which revealed a 34% reduction in the incidence of the primary endpoint in patients undergoing a percutaneous intervention compared to a 7% reduction in patients who did not (Figure 3). A second group of patients with remarkable benefits from GP IIb/IIIa receptor antagonist therapy include those with elevated troponin levels. This is evidenced by the impressive reduction of death in troponin positive patients by 42%, 67% and 70% in the PARAGON-B, PRISM46 and CAPTURE47 trials, respectively. Again, the meta-analysis supports this conclusion, and shows a 58% reduction in the 30-day incidence of death or myocardial infarction in the troponin positive group compared to a 5% increase in the troponin negative group (Figure 3). One other possible way to stratify patients is to use the TIMI risk score (to be described later) and administer GP IIb/IIIa receptor antagonists only in those high-risk patients (i.e., a score of 4 or greater). This strategy is supported by a retrospective analysis of the PRISM-PLUS study, whereby tirofiban was beneficial only in those high-risk patients, and no benefit was seen in others (Figure 4).48 One question to be answered is whether the observed efficacy between different compounds is due to pharmacological differences or merely a failure to achieve adequate levels of platelet inhibition. For example, in the TARGET trial,49 abciximab was shown to be superior to tirofiban at 30 days in patients with ACS undergoing percutaneous revascularization. Recent data, however, suggest that the tirofiban used in this trial failed to achieve > 80% platelet inhibition in the early stages of the infusion. Novel therapeutic approaches. New insights into the inflammatory component of ACS are leading to novel therapeutic approaches that go beyond the thrombotic process. Recently, the MIRACL trial (Figure 5)50,51 evaluated the effect of treatment with atorvastatin, 80 mg/day, initiated 24–96 hours after an ACS. The primary endpoint event, defined as death, nonfatal acute myocardial infarction, cardiac arrest with resuscitation or recurrent symptomatic myocardial ischemia, occurred in 14.8% in the atorvastatin group and 17.4% in the placebo group (p = 0.048). It is important to note that this beneficial effect was seen irrespective of baseline lipid levels, and may be related to the statin anti-inflammatory effects.52–54 Aspirin, initially thought as mainly an antiplatelet agent, is becoming more recognized for its anti-inflammatory properties. Clinical evidence to support this role includes its beneficial effect directly related to CRP levels.55 Preliminary data suggest that matrix metalloproteinase inhibitors can delay neointimal formation in various preparations, but more extensive work is still needed.56,57 Finally, ACE inhibitors have recently been demonstrated to possess potent anti-inflammatory properties58 that may explain some of their beneficial cardiovascular effects.59 See continuation of article in Part II
1. Constantinides P. Plaque fissuring in human coronary thrombosis. J Atheroscler Res 1966;6:1–6. 2. Davies MJ, Thomas AC. Plaque fissuring — The cause of acute myocardial infarction, sudden cardiac death, and crescendo angina. Br Heart J 1985;53:363–373. 3. Falk E. Plaque rupture with severe pre-existing stenosis precipitating coronary thrombosis. Characteristics of coronary atherosclerotic plaques underlying fatal occlusive thrombi. Br Heart J 1983;50:127–134. 4. Fuster V, Badimon L, Badimon JJ, Chesebro JH. The pathogenesis of coronary artery disease and the acute coronary syndromes. N Engl J Med 1992;326:242–250. 5. Libby P. Molecular bases of the acute coronary syndromes. Circulation 1995;91:2844–2850. 6. Tomassi S, Carluccio E, Bentivoglio M, et al. C-reactive protein as a marker of cardiac ischemic events in the year after a first, uncomplicated myocardial infarction. Am J Cardiol 1999;83:1595–1599. 7. Harb TS, Zareba W, Moss AJ, et al. Association of C-reactive protein and serum amyloid A with recurrent coronary events in stable patients after healing of acute myocardial infarction. Am J Cardiol 2002;89:216–221. 8. Ridker PM, Rifai N, Stampfer MJ, Hennekens CH. Plasma concentration of interleukin-6 and the risk of future myocardial infarction among apparently healthy men. Circulation 2000;101:1767–1772. 9. Biasucci LM, Liuzzo G, Caligiuri G, et al. Temporal relation between ischemic episodes and activation of the coagulation system in unstable angina. Circulation 1996;93:2121–2127. 10. Ma J, Hennekens CH, Ridker PM, Stampfer MJ. A prospective study of fibrinogen and risk of myocardial infarction in the Physicians’ Health Study. J Am Coll Cardiol 1999;33:1347–1352. 11. Amento EP, Ehsani N, Palmer H, Libby P. Cytokines positively and negatively regulate interstitial collagen gene expression in human vascular smooth muscle cells. Arteriosclerosis 1991;11:1223–1230. 12. Shah PK, Falk E, Badimon JJ, et al. Human monocyte-derived macrophages induce collagen breakdown in fibrous caps of atherosclerotic plaques. Potential role of matrix-degrading metalloproteinases and implications for plaque rupture. Circulation 1995;92:1565–1569. 13. Stefanadis C, Diamantopoulos L, Vlachopoulos C, et al. Thermal heterogeneity within human atherosclerotic coronary arteries detected in vivo: A new method of detection by application of a special thermography catheter. Circulation 1999;99:1965–1971. 14. MacIsaac AI, Thomas JD, Topol EJ. Toward the quiescent coronary plaque. J Am Coll Cardiol 1993;22:128–141. 15 Lewis HD Jr., Davis JW, Archibald DG, et al. Protective effects of aspirin against acute myocardial infarction and death in men with unstable angina: Results of the Veterans Administration Cooperative Study. N Engl J Med 1983;309:396–403. 16. Cairns JA, Gent M, Singer J, et al. Aspirin, sulfinpyrazone, or both in unstable angina. Results of a Canadian multicenter trial. N Engl J Med 1985;313:1369–1375. 17. Theroux P, Ouimet H, McCans J, et al. Aspirin, heparin, or both to treat acute unstable angina. N Engl J Med 1988;319:1105–1111. 18. RISC Group. Risk of myocardial infarction and death during treatment with low dose aspirin and intravenous heparin in men with unstable coronary artery disease. Lancet 1990;336:827–830. 19. Gum PA, Kottke-Marchant K, Poggio ED, et al. Profile and prevalence of aspirin resistance in patients with cardiovascular disease. Am J Cardiol 2001;88:230–235. 20. Eikelboom JW, Hirsh J, Weit JL, et al. Aspirin-resistant thromboxane biosynthesis and the risk of myocardial infarction, stroke, or cardiovascular death in patients at high risk for cardiovascular events. Circulation 2002;105:1650–1655. 21. CAPRIE Steering Committee. A randomized, blinded trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). Lancet 1996;348:1329–1339. 22. Bertrand ME, Rupprecht HJ, Gershlick AH, for the CLASSICS Investigators. Double-blind study of the safety of clopidogrel with and without a loading dose in combination with aspirin after coronary stenting: The clopidogrel aspirin stent international cooperative study (CLASSICS). Circulation 2000;102:624–629. 23. Mehta SR, Yusuf S. The Clopidogrel in Unstable Angina to Prevent Recurrent Events (CURE) Study Investigators. The CURE trial programme: Rationale, design and baseline characteristics including a meta-analysis of the effects of thienopyridines in vascular disease. Eur Heart J 2000;2:2033–2041. 24. The Clopidogrel in Unstable Angina to Prevent Recurrent Events Trial Investigators. Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation. N Engl J Med 2001;345:494–502. 25. Mehta SR, Yusuf S, Peters RJ, et al. Clopidogrel in Unstable Angina to Prevent Recurrent Events (CURE) Trial Investigators. Effects of pretreatment with clopidogrel and aspirin followed by long-term therapy in patients undergoing percutaneous coronary intervention: The PCI-CURE Study. Lancet 2001;358:527–533. 26. Cohen M, Adams PC, Parry G, et al., for the Antithrombotic Therapy in Acute Coronary Syndromes Research Group: Combination antithrombotic therapy in unstable rest angina and non-Q wave infarction non-prior aspirin users: Primary endpoints analysis from the ATACS trial. Circulation 1994;89:81–88. 27. Cohen M, Adams PC, Hawkins L, et al. Usefulness of antithrombotic therapy in resting angina pectoris or non-Q wave myocardial infarction in preventing death and myocardial infarction (A pilot study from the Antithrombotic Therapy in Acute Coronary Syndromes Study Group). Am J Cardiol 1990;66:1287–1292. 28. Holdright D, Patel D, Cunningham D, et al. Comparison of the effect of heparin and aspirin versus aspirin alone on transient myocardial ischemia and in-hospital prognosis in patients with unstable angina. J Am Coll Cardiol 1994;24:39–45. 29. Gurfinkel EP, Manos EJ, Mejail RI, et al. Low molecular weight heparin versus regular heparin or aspirin in the treatment of unstable angina and silent ischemia. J Am Coll Cardiol 1995;26:313–318. 30. Oler A, Whooley MA, Oler J, Grady D. Adding heparin to aspirin reduces the incidence of myocardial infarction and death in patients with unstable angina. A meta-analysis. JAMA 1996;276:811–815. 31. Cohen M, Demers C, Gurfinkel EP, et al. A comparison of low-molecular-weight heparin with unfractionated heparin for unstable coronary artery disease. N Engl J Med 1997;337:447–452. 32. Antman EM, McCabe CH, Gurfinkel EP, et al. Enoxaparin prevents death and cardiac ischaemic events in unstable angina/non-Q-wave myocardial infarction. Results of the Thrombolysis In Myocardial Infarction (TIMI) 11B trial. Circulation 1999;100:1593–1601. 33. Antman EM, Cohen M, Radley D, et al. Assessment of the treatment effect of enoxaparin for unstable angina/non-Q-wave myocardial infarction: TIMI 11B-ESSENCE meta-analysis. Circulation 1999;100:1602–1608. 34. Klein W, Buchwald A, Hillis SE, et al. Comparison of low-molecular-weight heparin with unfractionated heparin acutely and with placebo for 6 weeks in the management of unstable coronary artery disease. Fragmin in Unstable Coronary Artery Disease Study (FRIC). Circulation 1997;96:61–68. 35. Fragmin During Instability in Coronary Artery Disease (FRISC) study group: Low-molecular-weight heparin during instability in coronary artery disease. Lancet 1996;347:561–568. 36. FRISC II Investigators. Invasive compared with non-invasive treatment in unstable coronary-artery disease: FRISC II prospective randomized multicentre study. Fragmin and Fast Revascularization During Instability in Coronary Artery Disease Investigators. Lancet 1999;354:701–707. 37. FRISC II Investigators. Invasive compared with non-invasive treatment in unstable coronary artery disease: FRISC II prospective randomized multicentre study. Fragmin and Fast Revascularization During Instability in Coronary Artery Disease Investigators. Lancet 1999;354:708–715. 38. The FRAXIS Study Group: Comparison of two treatment durations (6 days and 14 days) of a low molecular weight heparin with a 6-day treatment of unfractionated heparin in the initial management of unstable angina or non-Q wave myocardial infarction: FRAXIS (FRAXiparine in Ischemic Syndrome). Eur Heart J 1999;20:1553–1562. 39. PRISM Study Investigators. A comparison of aspirin plus tirofiban with aspirin plus heparin for unstable angina. N Engl J Med 1998;338:1498–1505. 40. PRISM-PLUS Study Investigators. Inhibition of the platelet glycoprotein IIb/IIIa receptor with tirofiban in unstable angina and non-Q wave myocardial infarction. N Engl J Med 1998;338:1488–1497. 41. PURSUIT Trial Investigators. Inhibition of platelet glycoprotein IIb/IIIa with eptifibatide in patients with acute coronary syndromes. N Engl J Med 1998;339:436–443. 42. PARAGON Investigators. International, randomized, controlled trial of lamifiban (a platelet glycoprotein IIb/IIIa inhibitor, heparin, or both in unstable angina). Circulation 1998;97:2386–2395. 43. GUSTO IV-ACS Investigators. Effect of glycoprotein IIb/IIIa receptor blocker abciximab on outcome in patients with acute coronary syndromes without early coronary revascularization: The GUSTO IV-ACS randomized trial. Lancet 2001;357:1915–1924. 44. Chew DP, Moliterno DJ. A critical appraisal of platelet glycoprotein IIb/IIIa inhibition. J Am Coll Cardiol 2000;36:2028–2035. 45. Kleiman NS, Lincoff AM, Flaker GC, et al. Early percutaneous coronary intervention, platelet inhibition with eptifibatide and clinical outcomes in patients with acute coronary syndromes. Circulation 2000;101:751–757. 46. Heeschen C, Hamm CW, Goldmann B, et al., for the PRISM Study Investigators. Troponin concentrations for stratification of patients with acute coronary syndromes on relation to therapeutic efficacy of tirofiban. Platelet Receptor Inhibition in Ischemic Syndrome Management (PRISM). Lancet 1999;354:1757–1762. 47. CAPTURE Investigators. Randomised placebo-controlled trial of abciximab before and during coronary intervention in refractory unstable angina: The CAPTURE study. Lancet 1997;349:1429–1435. 48. Morrow DA, Antman EM, Snapinn SM, et al. An integrated clinical approach to predicting the benefit of tirofiban in non-ST elevation acute coronary syndromes. Application of the TIMI Risk Score for UA/NSTEMI in PRISM-PLUS. Eur Heart J 2002;23:223–229. 49. Topol EJ, Moliterno DJ, Herrmann HC. Comparison of two platelet glycoprotein IIb/IIIa inhibitors, tirofiban and abciximab, for the prevention of ischemic events with percutaneous coronary revascularization. N Engl J Med 2001;344:1888–1894. 50. Schwartz GC, Oliver MF, Ezekowitz MD, et al. Rationale and design of the Myocardial Ischemia Reduction with Aggressive Cholesterol Lowering (MIRACL) study that evaluates atorvastatin in unstable angina pectoris and in non-Q wave myocardial infarction. Am J Cardiol 1998;81:578–581. 51. Schwartz GG, Olsson AG, Ezekowitz MD, et al. Effects of atorvastatin on early recurrent ischemic events in acute coronary syndromes: The MIRACL study: A randomized controlled trial. Myocardial Ischemia Reduction with Aggressive Cholesterol Lowering (MIRACL) Study Investigators. JAMA 2001;285:1711–1718. 52. Crisby M, Nordin-Fredriksson G, Shah PK, et al. Pravastatin treatment increases collagen content and decreases lipid content, inflammation, metalloproteinases, and cell death in human carotid plaques: Implications for plaque stabilization. Circulation 2001;103:926–933. 53. Albert MA, Danielson E, Rifai N, Ridker PM, for the PRINCE Investigators. Effect of statin therapy on C-reactive protein levels: The pravastatin inflammation/CRP evaluation (PRINCE): A randomized trial and cohort study. JAMA 2001;286:64–70. 54. Ridker PM, Rifai N, Pfeffer MA, et al. Long-term effects of pravastatin on plasma concentration of C-reactive protein. The Cholesterol and Recurrent Events (CARE) Investigators. Circulation 1999;100:230–235. 55. Ridker PM, Cushman M, Stampfer MJ, et al. Inflammation, aspirin, and the risk of cardiovascular disease in apparently healthy men. N Engl J Med 1997;336:973–979. 56. Damas JK, Waehre T, Yndestad A, et al. Stromal cell-derived factor-1alpha in unstable angina: Potential antiinflammatory and matrix-stabilizing effects. Circulation 2002;106:36–42. 57. Herman MP, Sukhova GK, Kisiel W, et al. Tissue factor pathway inhibitor-2 is a novel inhibitor of matrix metalloproteinases with implications for atherosclerosis. J Clin Invest 2001;107:1117–1126. 58. Ambrose AJ, Martinez EE. A new paradigm for plaque stabilization. Circulation 2002;105:2000–2004. 59. Yusuf S, Sleight P, Pogue J, et al. Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high risk patients. The Heart Outcomes Prevention Evaluation Study Investigators. N Engl J Med 2000;34:145–153. 60. The TIMI-IIIB Investigators. Effects of tissue plasminogen activator and a comparison of early invasive and conservative strategies in unstable angina and non-Q wave myocardial infarction: Results of the TIMI-IIIB trial. Circulation 1994;89:1545–1556. 61. Ferry DR, O’Rourke RA, Blaustein AS, et al. Design and baseline characteristics of the Veterans Affairs Non-Q Wave Infarction Strategies In-Hospital (VANQWISH) trial. VANQWISH Trial Research Investigators. J Am Coll Cardiol 1998;31:312–320. 62. Boden WE, O’Rourke RA, Crawford MH, et al. Outcomes in patients with acute non-Q-wave myocardial infarction randomly assigned to an invasive as compared with a conservative management strategy. Veterans Affairs Non-Q Wave Infarction Strategies in Hospital (VANQWISH) trial investigators. N Engl J Med 1998;338:1785–1792. 63. McCullough PA, O’Neill WW, Graham M, et al. A prospective randomized trial of triage angiography in acute coronary syndromes ineligible for thrombolytic therapy. Results of the medicine versus angiography in thrombolytic exclusion trial (MATE) trial. J Am Coll Cardiol 1998;32:596–605. 64. FRISC II Investigators. Invasive compared with non-invasive treatment in unstable coronary-artery disease: FRISC II prospective randomized multicentre study. Fragmin and Fast Revascularization During Instability in Coronary Artery Disease Investigators. Lancet 1999;354:708–715. 65. Wallentin L, Lagerqvist B, Husted S, et al. Outcomes at one year after an invasive compared with a non-invasive strategy in unstable coronary-artery disease. The FRISC II invasive randomized trial. FRISC II Investigators. Fast Revascularization During Instability in Coronary Artery Disease. Lancet 2000;356:9–16. 66. Cannon CP, Weintraub WS, Demopoulos LA, et al. TACTICS (Treat Angina with Aggrastat and Determine Cost of Therapy with an Invasive or Conservative Strategy). Thrombolysis in Myocardial Infarction 18 Investigators. Comparison of early invasive and conservative strategies in patients with unstable coronary syndromes treated with the glycoprotein IIb/IIIa inhibitor tirofiban. N Engl J Med 2001;344:1879–1887. 67. Antman EM, Cohen M, Bernink PJ, et al. The TIMI risk score for unstable angina/non-ST elevation MI: A method for prognostication and therapeutic decision-making. JAMA 2000;284:835–842.

Advertisement

Advertisement

Advertisement