Skip to main content

Advertisement

ADVERTISEMENT

Feature

Overcoming Heparin Limitations in High-Risk Percutaneous Coronary Intervention: The Alternative Strategy — Replacing Heparin wit

Timothy D. Henry, MD
April 2002
Adverse outcomes of PCI include myocardial ischemia and necrosis, the need for endovascular or surgical reintervention, and occasionally death.1–3 Routine pharmacologic therapy includes antiplatelet and antithrombotic agents. Heparin has been the standard antithrombin, but has a narrow therapeutic window and significant limitations.4 In recent years, the pharmacological and clinical limitations of heparin in PCI have been addressed by adding more potent antithrombin agents including GP IIb/IIIa inhibitors and thienopyridines.5,6 This add-on strategy has reduced ischemic complication rates by 30–50% relative to low-dose, weight-adjusted heparin. However, the incidence of clinically significant bleeding (transfusion, TIMI major bleeding and minor bleeding) has increased almost universally. Furthermore, practice series indicate that both ischemic and bleeding complications remain a substantial clinical and economic problem among the general PCI population.7 A potentially attractive alternative strategy is to first overcome the limitations of heparin with more effective and safer antithrombin agents, and then determine the need for potent antiplatelet agents as needed in appropriate patients. Heparin, the historical foundation antithrombin Unfractionated heparin has been the standard intravenous antithrombin therapy in PCI from the outset. Pioneering work used heparin at doses of 3,000 U boluses, but with dextran and prolonged warfarin co-medication.8 By the early 1990s, data from case-control studies suggested that in the presence of aspirin, doses of heparin in the range of 10,000–15,000 U targeting activated clotting times (ACT) of 300–350 seconds (Hemochron) were more effective than lower doses,9–11 but were not associated with increased bleeding risk unless infusions were prolonged after PCI.12 With the introduction of glycoprotein IIb/IIIa inhibitors, bleeding risk was increased. In response, doses of heparin were reduced, target ACT levels were maintained at 200–250 seconds and vascular access site management was amended.13,14 How good is heparin — especially in high-risk patients? Formal studies of heparin in PCI have been surprisingly infrequent and heparin treatment remains empiric. But since heparin is ubiquitous in PCI, the reported data prior to the use of GP IIb/IIIa inhibitors provide a basis on which to judge the performance of this indirect antithrombin agent. Risk factors for clinical ischemic complications of PCI are summarized in Table 1. Heparin efficacy: High-risk demographic characteristics. Higher adverse event rates among certain patient groups undergoing angioplasty with heparin alone form the basis for advocating alternative treatment strategies. Specifically, co-investigators of the NHLBI angioplasty registry2,15 and others2,16–21 have demonstrated increased risk of death and myocardial ischemia in elderly patients treated with heparin for PCI, with some question surrounding outcomes in octogenarians.3,17,22 Likewise, female gender has been implicated as an independent risk factor in some series,23,24 but not others,2,25–27 while low-normal (2) and low (2) or high (> 35 kg/m2) body mass index appears to increase the risk of death 2–7-fold.28 Interestingly, age, gender and low body weight are also powerful drivers of bleeding risk from heparin in PCI and are often coexistent.29,30Heparin efficacy: High-risk clinical characteristics. Diabetes mellitus is strongly associated with coronary artery disease31,32 and reported in approximately 20% of patients undergoing PCI in clinical trials.14,33–40 Diabetes predicts increased risk of restenosis41 and may increase the rate of death, MI and urgent revascularization, as observed in the EPISTENT study.34 This increased risk is probably explained by an association of diabetes with increased incidence of plaque ulceration and increased potential for intracoronary thrombus formation. Angioscopic studies in diabetic and non-diabetic patients with unstable angina have revealed ulcerated plaque in 94% of the diabetics versus 60% in non-diabetics (p = 0.01) and intracoronary thrombi was seen in 94% of diabetics versus 55% of non-diabetics (p = 0.004).31,42 Intracoronary thrombus at the time of PCI therefore seems to be an important factor in the poor outcomes of diabetic patients.3 Renal function has emerged as an important independent predictor of mortality and other complications of coronary artery disease.43,44 The prevalence of chronic renal disease (CRD) is growing at 7% per year in the United States,45 but the incidence is underestimated by serum creatinine so that calculation or measurement of creatinine clearance is a necessary step in patient evaluation. Data from three trials of patients with ACS suggested that the prevalence of renal impairment (defined as creatinine clearance 46 this has been confirmed in the PCI population, where 40% of patients have creatinine clearance 8,47 Chronic renal disease predicts a 2-fold increase in mortality following coronary artery bypass surgery.48 PCI results may be even worse;49–51 even mild renal insufficiency is independently associated with increased mortality.7 Mechanisms underlying increased risk in patients with CRD have not been completely defined. Associated risk factors such as diabetes, lipid abnormalities and hypertension may contribute, but this confounding seems to be ruled out by multivariate analyses identifying renal function as an independent risk factor.7,47 Patients with CRD have more extensive vascular disease and more complex coronary anatomy.51 Chronic renal disease may create an increased thrombotic tendency independently, mediated by elevated abnormalities in platelet function and activation of the coagulation system,52–58 which potentiates the risk associated with increased lesion complexity or patient demography. Finally, these patients have an increased risk of bleeding complications as well. ACS patients, i.e., those presenting with unstable angina and non-ST elevated MI, were first classified by Braunwald in 1989 to aid in the decision regarding diagnostic measures and therapy of individual patients, and to provide a more precise basis for including patients in clinical trials, as well as evaluating the outcomes of these trials.59 In a subsequent consensus document, patients who failed to respond to heparin within 30 minutes were considered at increased risk of MI or cardiac death.60 The classification has been validated prospectively.61,62 DeFeyter et al.63 proposed adaptations of the Braunwald classification to improve the assessment of patients with unstable angina undergoing PCI. They reviewed 30 published PCI reports of patients treated with heparin, aspirin and PCI (no GP IIb/IIIa inhibitors or thienopyridines were used in these trials), thus representing a reasonable estimate of heparin effectiveness among these patients. Results of the analyses are summarized in Figure 1; they demonstrate the increased risk of ischemic outcomes after PCI in all three “high-risk” clinical presentations. Based on this analysis and observations made in a series of patients treated in Rotterdam, De Feyter proposed the risk stratification of unstable PCI patients (Table 2). The mechanism of increased risk for heparin in UA patients is almost certainly the result of intracoronary thrombus. This link is strongly established by angioscopy, which is a more sensitive method of detection for intracoronary thrombus than angiography. Ramee et al.31,64 first reported that thrombus was observed in 15/16 UA patients (94%) by angioscopy but only 2/16 patients (13%) using angiography. In a larger, multicenter series reported by the same authors,31,65 a total of 122 patients undergoing PCI were subjected to angiography and angioscopy. Ninety-five patients (78%) had unstable angina and 27 patients (22%) were stable. Once again, angioscopy (61% incidence) was more sensitive at detecting thrombus than angiography (20%; p 11 reported that patients who experienced adverse clinical outcomes after PCI had significantly lower initial and minimum median ACT times. The odds ratio for adverse outcomes among patients with ACT >= 300 seconds and 9 (0.23; 95% CI, 0.15–0.37) and McGarry et al.10 (0.22; 95% CI, 0.08–0.65). The combined odds ratio for death or ischemic complications in cases among all three studies was 0.25 (95% CI, 0.17–0.37; p 71 The report by Narins et al. also included a multivariable binary logistic regression model that predicted probability of ischemic complications at any given ACT level. The relation between the initial ACT and adverse outcomes was highly statistically significant (p = 0.015). Moving from the 25th percentile of ACT (324 seconds) to the 75th percentile (413 seconds), the probability of adverse ischemic outcomes declined from 7.9% to 4.5%. These data (published in 1996) are strikingly similar to those reported by Chew et al. in 2001 using aggregate data from 6 randomized trials in 5,216 PCI patients. In this report, an ACT of 350–375 seconds was associated with 6.6% incidence of death, MI or revascularization, compared to 10.1% for an ACT range of 171–295 seconds (p 4 These data support the view that the dose and the pharmacodynamic response to heparin are key determinants in the risks associated with PCI. The best results are seen when the ACT reaches 350 seconds or more. Below this, event rates on heparin are high, as shown in the EPISTENT and ESPRIT trials, in which stents were deployed in elective, low-risk patients. The EPISTENT trial34 reported 10.8% incidence of the so-called triple endpoint, and the ESPRIT trial37 reported 10.5% among elective PCI patients (Table 3). Whether current advances demonstrate a similar magnitude of benefit when compared with higher levels of heparin anticoagulation has not been determined. Unfortunately, this conjecture cannot be adequately put to the test. The only two published randomized trials of heparin dosing have not solved this question due to small samples sizes. Vainer et al.72 reported 13.2% ischemic complications with 5,000 U heparin (ACT, 209 seconds) compared to 8.0% with 20,000 U (ACT, 381 ± 117 seconds; odds ratio, 1.74; 95% CI, 0.91–3.35). Conversely, Boccara et al.73 reported freedom from death, MI, unplanned revascularization or bail-out stenting in 91% of patients treated with 15,000 U and 95% treated with 100 U/kg (odds ratio, 1.88; 95% CI, 0.80–4.50). The conflicting and inconclusive nature of these data may be the result of small sample sizes. The smoking gun of PCI complications: Intracoronary thrombus In an era of frequent and extensive stenting, increased ischemic risk can largely be attributed to an elevated thrombotic potential. As reviewed, demographic risk factors are associated with procoagulant states; diabetes and chronic kidney diseases are also associated with arterial pathology leading to surface disruption and increased likelihood of thrombus formation. Unstable coronary syndromes are associated with increased incidence of coronary thrombus and systemic heparin resistance. Finally, even anatomic complexity is perhaps a marker for often undetected red clot in the artery. Why does heparin fail when thrombus is present? Unfractionated heparin is a variable chain-length glycosaminoglycan. It acts indirectly by accelerating conformational changes in antithrombin (AT) that enable inhibition of the clotting factors Xa and thrombin.74,75 Thrombin is also the key plasma protease responsible for cleavage of fibrinogen to fibrin, and activation of a broad range of critical platelet functions.76,77 Unfortunately, heparin has pharmacokinetic and biophysical limitations that are dependent on its structure and mechanism of action and these limitations become clear in patients with risk factors for intracoronary thrombosis and ischemic complications in PCI. First, heparin’s non-specific binding to endothelium and to plasma proteins, such as vitronectin, fibronectin, histidine-rich glycoprotein, platelet factor-IV, and high molecular weight multimers of von Willebrand factor result in variable pharmacodynamic effects in patients with various clinical presentations.78 Binding to cells and plasma proteins decreases the anticoagulant effect of heparin by limiting its availability to interact with AT.79 In addition, platelet factor-IV, a highly cationic protein that binds heparin with high affinity, and high molecular weight moieties of von Willebrand factor are released by activated platelets during clotting.80 Furthermore, some clinical studies have demonstrated that heparin increases the likelihood of platelet aggregation in ACS patients.81 Perhaps the most significant limitation of heparin for management of high-risk PCI patients lies in its inability to inhibit thrombin bound to fibrin.82 In addition, factor Xa bound to the surface of activated platelets is also resistant to inactivation by the heparin:AT complex83,84 and, as part of the prothrombinase complex, converts prothrombin to thrombin, thereby increasing the amount of thrombin available to bind to fibrin. Furthermore, thrombin binds to fibrin via exosite-1, a substrate-binding site that is distinct from thrombin’s active site.85–87 Current evidence suggests that fibrin-bound thrombin is protected from inactivation by the heparin:AT complex because heparin binds simultaneously to fibrin and to the heparin-binding domain on thrombin, so-called exosite-2, thereby bridging thrombin onto fibrin.88 The formation of a ternary heparin:thrombin:fibrin complex increases the affinity of thrombin for fibrin and induces conformational changes in the active site of thrombin. The resistance of thrombin within the ternary complex to inactivation by fluid-phase inhibitors may reflect allosteric modulation of its active site or spacial constraints that impair the reactivity of inhibitors with thrombin.89,90 Many of the limitations of unfractionated heparin with respect to clot-bound thrombin also apply to low molecular weight heparins — regardless of Xa:IIa ratios.91 Continued on next page
1. Mahaffey KW, Roe MT, Dyke CK, et al. Misreporting of myocardial infarction end points: Results of adjudication by a central clinical events committee in the PARAGON-B trial. Second Platelet IIb/IIIa Antagonist for the Reduction of Acute Coronary Syndrome Events in a Global Organization Network Trial. Am Heart J 2002;143:242–248. 2. Ellis SG. Elective coronary angioplasty: Techniques and complications. In: Topol E (ed). Textbook of Interventional Cardiology. WB Saunders: Philadelphia, 1999: pp. 147–162. 3. Lincoff AM, Topol E. Abrupt vessel closure. In: Topol E (ed). Textbook of Interventional Cardiology. WB Saunders: Philadelphia, 1999: pp. 163–187. 4. Chew DP, Bhatt DL, Lincoff AM, et al. Defining the optimal activated clotting time during percutaneous coronary intervention: Aggregate results from 6 randomized, controlled trials. Circulation 2001;103:961–966. 5. Mehta SR, Eikelboom JW, Rupprecht HJ, et al. 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. 6. Leon MB, Baim DS, Popma JJ, et al. A clinical trial comparing three antithrombotic drug regimens after coronary stenting. Stent Anticoagulation Restenosis Study Investigators. N Engl J Med 1998;339:1665–1671. 7. Best P, Lennon R, Ting H, et al. Even mild renal insufficiency is associated with increased mortality after percutaneous coronary intervention. J Am Coll Cardiol 2001;37:76A. 8. Gruentzig AR, Senning A, Siegenthaler WE. Nonoperative dilatation of coronary-artery stenosis: Percutaneous transluminal coronary angioplasty. N Engl J Med 1979;301:61–68. 9. Ferguson JJ, Dougherty KG, Gaos CM, et al. Relation between procedural activated coagulation time and outcome after percutaneous transluminal coronary angioplasty. J Am Coll Cardiol 1994;23:1061–1065. 10. McGarry TF Jr., Gottlieb CR, Morganroth J, et al. The relationship of anticoagulation level and complications after successful percutaneous transluminal coronary angioplasty. Am Heart J 1992;123:1445–1451. 11. Narins CR, Hillegass WB Jr., Nelson CL, et al. Relation between activated clotting time during angioplasty and abrupt closure. Circulation 1996;93:667–671. 12. Kong DF, Califf RM. Post-procedure heparin: Boon or burden? (editorial; comment). Am Heart J 1998;136:183–185. 13. The PROLOG Investigators. Standard versus low-dose weight-adjusted heparin in patients treated with the platelet glycoprotein IIb/IIIa receptor antibody fragment abciximab (c7E3 Fab) during percutaneous coronary revascularization. Am J Cardiol 1997;79:286–291. 14. The EPILOG Investigators. Platelet glycoprotein IIb/IIIa receptor blockade and low-dose heparin during percutaneous coronary revascularization (see comments). N Engl J Med 1997;336:1689–1696. 15. Holmes DR, Holubkov R, Vlietstra RE, et al. Co-investigators of the National Heart, Lung and Blood Institute Percutaneous Transluminal Coronary Angioplasty Registry: Comparison of complications during percutaneous transluminal coronary angioplasty from 1977–1981 and from 1985–1986: The National Heart, Lung, and Blood Institute Percutaneous Transluminal Coronary Angioplasty Registry. J Am Coll Cardiol 1988;12:1149–1155. 16. Simpfendorfer C, Raymond R, Schraider J, et al. Early and long term results of percutaneous transluminal coronary angioplasty in patients 70 years of age and older with angina pectoris. Am J Cardiol 1988;62:959–961. 17. Kern MJ, Deligonul U, Galan K, et al. Percutaneous transluminal coronary angioplasty in octogenarians. Am J Cardiol 1988;61:457–458. 18. de Jaegere PP, De Feyter P, van Domburg R, et al. Immediate and long term results of percutaneous transluminal coronary angioplasty in patients aged 70 and over. Br Heart J 1992;67:138–142. 19. Thompson RC, Holmes DR Jr., Gersh BJ, et al. Percutaneous transluminal coronary angioplasty in the elderly: Early and long term results. J Am Coll Cardiol 1991;17:1245–1250. 20. ten Berg J, Voors MM, Suttorp MJ, et al. Long term results after successful percutaneous transluminal coronary angioplasty in patients over 75 years of age. Am J Cardiol 1996;77:690–695. 21. Tan K, Sulke N, Taub N, et al. Percutaneous transluminal coronary angioplasty in patients 70 years of age or older: 12 years’ experience. Br Heart J 1995;74:310–317. 22. Jeroudi M, Kleiman NS, Minor ST, et al. Percutaneous transluminal coronary angioplasty in octogenarians. Ann Intern Med 1990;113:423–428. 23. Cowley M, Dorros G, Kelsey SF. Acute coronary events associated with percutaneous transluminal coronary angioplasty. Am J Cardiol 1983;53:12C–16C. 24. Ellis S, Roubin GS, King SB, et al. Angiographic and clinical predictors of acute closure after native vessel coronary angioplasty. Circulation 1988;77:372–379. 25. McEniery PT, Hollman J, Knezinek V, et al. Comparative safety and efficacy of percutaneous transluminal coronary angioplasty in men and women. Cathet Cardiovasc Diagn 1987;13:364–371. 26. Kelsey SF, James M, Holubkov AL, et al. Results of percutaneous transluminal coronary angioplasty in women: 1985–1986 National Heart, Lung, and Blood Institute’s Coronary Angioplasty Registry. Circulation 1993;87:720–727. 27. Weintraub WS, Wenger NK, Kosinski AS, et al. Percutaneous transluminal coronary angioplasty in women compared with men. J Am Coll Cardiol 1994;24:81–90. 28. Ellis S, Elliott J, Horrigan M, et al. Low-normal or excessive body mass index: Newly identified and powerful risk factors for death and other complications with percutaneous coronary intervention. Am J Cardiol 1996;78:642–646. 29. Aguirre FV, Topol EJ, Ferguson JJ, et al. Bleeding complications with the chimeric antibody to platelet glycoprotein IIb/IIIa integrin in patients undergoing percutaneous coronary intervention. EPIC Investigators. Circulation 1995;91:2882–2890. 30. Feit F, Bittl JA, Keller NM, et al. Hemorrhagic complications in association with percutaneous coronary intervention: Can the risk be attenuated? J Invas Cardiol 2000;12(Suppl F):7F–13F. 31. White CJ, Ramee SR. Coronary angioscopy. In: Topol E (ed). Textbook of Interventional Cardiology. WB Saunders Company: Philadelphia, 1999: pp. 783–800. 32. Usitupa M, Siitonen O, Aro A. Prevalence of coronary heart disease, left ventricular failure, and hypertension in middle-aged, newly diagnosed type 2 (non-insulin dependent) diabetic subjects. Diabetologia 1985;28:22. 33. The EPIC Investigators. Use of monoclonal antibody directed against the platelet glycoprotein IIb/IIIa receptor in high-risk coronary angioplasty. N Engl J Med 1994;330:956–961. 34. The EPISTENT Investigators. Randomized, placebo-controlled and balloon-angioplasty-controlled trial to assess safety of coronary stenting with use of platelet glycoprotein IIb/IIIa blockade. Evaluation of Platelet IIb/IIIa Inhibitor for Stenting. Lancet 1998;352:87–92. 35. The CAPTURE Investigators. Randomized placebo-controlled trial of abciximab before and during coronary intervention in refractory unstable angina: The CAPTURE study. Lancet 1997;349:1429–1435. 36. The IMPACT-II Investigators. Randomized placebo-controlled trial of effect of eptifibatide on complications of percutaneous coronary intervention: IMPACT-II. Lancet 1997;348:1422–1428. 37. The ESPRIT Investigators. Novel dosing regimen of eptifibatide in planned coronary stent implantation (ESPRIT): A randomized, placebo-controlled trial. Lancet 2000;356:2037–2044. 38. The RESTORE Investigators. Effects of platelet glycoprotein IIb/IIIa blockade with tirofiban on adverse cardiac events in patients with unstable angina or acute myocardial infarction undergoing coronary angioplasty. Circulation 1997;96:1445–1453. 39. Topol EJ, Moliterno DJ, Hermann HC, et al. Comparison of two glycoprotein IIb/IIIa inhibitors, tirofiban and abciximab, for the prevention of ischemic events with percutaneous revascularization. N Engl J Med 2001;344:1888–1894. 40. Bittl JA, Chaitman B, Feit F, et al. Bivalirudin versus heparin during coronary angioplasty for unstable or post-infarction angina: The first reanalysis of the bivalirudin angioplasty study. Am Heart J 2001;142:952–959. 41. Miller JM, Ohman EM, Moliterno DJ, et al. Restenosis: The clinical issues. In: Topol EJ (ed). Textbook of Interventional Cardiology. WB Saunders Company: Philadelphia, 1999: pp. 379–415. 42. Silva J, Escobar A, Collins TJ, et al. Unstable angina: A comparison of angioscopic findings between diabetic and nondiabetic patients. Circulation 1995;92:1731–1736. 43. Redden DN, Szczech LA, Tuttle RH, et al. Impact of renal disease and treatment type on mortality in patients with clinically significant coronary artery disease (Abstr). Irish Nephrological Society Meeting 2001. 44. Herzog C, Ma J, Collins A. Long-term outcome of dialysis patients in the United States with coronary revascularization procedures. Kidney Int 1999;56:324–332. 45. USRDS 2000 Annual Data Report. National Institute of Health, National Institute of Diabetes, Digestive and Kidney Diseases: Bethesda, Maryland. 46. Al Suwaidi J, Reddan DN, Pieper KS, et al. Prognostic importance of abnormal renal function in patients with acute coronary syndromes. Circulation 2000;102:A3828. 47. Robson R. The use of bivalirudin in patients with renal impairment. J Invas Cardiol 2000;12(Suppl F):33F–36F. 48. Anderson RJ, O’Brien M, MaWhinney S, et al. Renal failure predisposes patients to adverse outcomes after coronary artery bypass surgery. VA Cooperative Study #5. Kidney Int 1999;55:1057–1062. 49. Gradaus F, Schoebel FC, Ivens K, et al. Rate of restenosis after PTCA in patients with terminal renal failure. A quantitative coronary angioplasty study. Z Kardiol 1997;86:373–379. 50. Rubenstein MH, Harrell LC, Sheynberg BV, et al. Are patients with renal failure good candidates for percutaneous coronary revascularization? Circulation 2000;102:2966–2972. 51. Asinger RW, Henry HD, Herzog CA, et al. Clinical outcomes of PTCA in chronic renal failure: A case-control study for comorbid features and evaluation of dialysis dependence. J Invas Cardiol 2001;13:21–28. 52. Remuzzi G. Bleeding in renal failure. Lancet 1988;1:1205–1208. 53. Sagripanti A, Barsotti G. Bleeding and thrombosis in chronic uremia. Nephron 1997;75:125–139. 54. Irish AB, Green FR. Factor VII coagulant activity (VIIc) and hypercoagulability in chronic renal disease and dialysis: Relationship with dyslipidemia, inflammation, and factor VII genotype. Nephrol Dial Transplant 1998;13:679–684. 54. Tomura S, Nakamura Y, Deguchi F, et al. Coagulation and fibrinolysis in patients with chronic renal failure undergoing conservative treatment. Thromb Res 1991;64:81–90. 56. Al-Saady NM, Leatham EW, Gupta S, et al. Monocyte expression of tissue factor and adhesion molecules: The link with accelerated coronary artery disease in patients with chronic renal failure. Heart 1999;81:134–140.

Advertisement

Advertisement

Advertisement