Article

Bleeding as a Prognostic Marker in Acute Coronary Syndromes

Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Average (ratings)
No ratings
Your rating
Copyright Statement:

The copyright in this work belongs to Radcliffe Medical Media. Only articles clearly marked with the CC BY-NC logo are published with the Creative Commons by Attribution Licence. The CC BY-NC option was not available for Radcliffe journals before 1 January 2019. Articles marked ‘Open Access’ but not marked ‘CC BY-NC’ are made freely accessible at the time of publication but are subject to standard copyright law regarding reproduction and distribution. Permission is required for reuse of this content.

There has been a significant evolution in the management of patients with acute coronary syndromes (ACS), largely driven by advances in pharmacological therapy. Increased implementation of evidence-based antithrombotics and antiplatelet agents has led to improved patient outcomes.1,2 Additionally, management algorithms favouring early invasive intervention in high-risk patients are gaining support.3,4 Despite improvements in endpoints such as death, myocardial infarction (MI) and recurrent ischaemia, these therapies are not without their risks, namely bleeding and blood transfusion. Recent studies suggest that bleeding is independently associated with an increased risk of both short- and longer-term adverse events (including MI, stroke and death) among patients with ACS.5-9 Furthermore, the safety of blood transfusion in this population has also been called into question,10-12 suggesting that management strategies that maintain an adequate anticoagulant effect to reduce ischaemia, while at the same time minimising the risk of bleeding, may further improve ACS outcomes.

Aetiology and Risk Factors for Bleeding in ACS

Rates of major bleeding in clinical studies of ACS have ranged from 0.8% to 11.5%5,6,13,14 and, according to some registries, it is the most common non-cardiac complication of therapy in this patient population.5,13 Clinical risk factors for in-hospital bleeding have been identified in analyses of several contemporary ACS trials and registry databases. Older age, female sex, lower body weight and chronic kidney disease have been consistently associated with an increased risk for bleeding complications. In addition, the use of anticoagulants, fibrinolytics and invasive procedures (e.g., cardiac catheterisation with or without intervention, intra-aortic counterpulsation) were also predictors of in-hospital bleeding. The most common sites of bleeding were vascular access sites and the gastrointestinal tract, followed by less common retroperitoneal bleeds and genito-urinary haemorrhage.5-7

There are also data to suggest that inappropriate dosing of anticoagulant medications is associated with bleeding in patients with ACS. In the Can Rapid Risk Stratification of Unstable Angina Patients Suppress Adverse Outcomes with Early Implementation of the ACC/AHA Guidelines (CRUSADE) registry, for instance, over 42% of all patients with non-ST-segment elevation (NSTE) ACS received at least one antithrombotic drug in excess.13

Not only were many of these medications overdosed, but a large number of these dosing errors occurred in vulnerable patients with risk factors for bleeding. In particular, dose adjustment of renally excreted anticoagulants, such as enoxaparin and eptifibatide, in patients with chronic kidney disease is critically important to reduce the risk of over-dosage and subsequent bleeding.15

Defining Clinical Bleeding Events

One of the more difficult barriers to overcome in clinical studies addressing bleeding in ACS has been the development of a consensus classification for bleeding severity. For example, the Global Use of Strategies to Open Occluded Coronary Arteries16 (GUSTO) investigators classified bleeding, using clinical variables, as either mild, moderate or severe/life-threatening. GUSTO severe or life-threatening bleeding is defined as either intracranial haemorrhage or bleeding resulting in haemodynamic compromise necessitating intervention. GUSTO moderate bleeding is defined as bleeding requiring transfusion, but not resulting in haemodynamic compromise. All other bleeding events are classified as GUSTO mild.

Alternatively, the Thrombolysis in Myocardial Infarction (TIMI)17 investigators utilised a more laboratory-based classification scheme when defining bleeding events. TIMI major bleeding is defined as intracranial haemorrhage or bleeding with a haemoglobin decrease of >5 g/dl or haematocrit decrease of >15%. The definition of TIMI minor bleeding is dependent on whether or not there is an identifiable source of blood loss. If a bleeding site is found, then TIMI minor bleeding is defined as a haemoglobin decrease of >3 g/dl or a haematocrit decrease of >10%; if no site is found, then it is defined as a haemoglobin decrease of >4 g/dl or haematocrit decrease of >12%. Finally, TIMI minimal bleeding is defined as any clinically overt sign of haemorrhage that is associated with a haemoglobin decrease of <3 g/dl or a haematocrit decrease of <9%.

While both of these definitions were developed in the context of thrombolytic therapy for ST-segment elevation MI (STEMI), they have been used extensively to classify bleeding events in trials and registries of non-ST-segment elevation (NSTE) ACS. However, no standard bleeding definition exists and other clinical trials in ACS have utilised a combination of the TIMI and GUSTO classifications or their own unique definitions.18.19Table 1 displays several of the commonly employed bleeding classification schemes used in clinical trials and registries of ACS.

Bleeding and Prognosis in ACS

Rather than merely a nuisance of therapy, bleeding in the setting of ACS has consistently been shown to be associated with adverse outcomes. Although the exact mechanisms underlying this risk association are unclear, it is likely that several factors contribute to the increased mortality seen in these studies. First, bleeding complications likely lead to cessation or reversal of anticoagulation that may precipitate rebound ischaemia and/or recurrent infarction. Second, several human and animal models of bleeding and haemorrhagic shock have shown upregulation of inflammatory cytokines, vasoactive substances and modulators of the neurohormonal system, all of which help to maintain vital organ perfusion, but all of which have also been linked to adverse cardiovascular outcomes.20-22 Third, chronic bleeding may lead to anaemia, which is independently associated with increased mortality in patients with coronary artery disease (see below). Finally, administration of blood transfusion may also serve to increase the risk of ischaemic events and subsequent mortality (see below).

While the pathophysiology linking bleeding with adverse cardiovascular events in ACS has been poorly defined, the association between bleeding and prognosis has been demonstrated in several clinical studies. Major bleeding in the large, multicentre, prospective Global Registry of Acute Coronary Events (GRACE)5 registry was found to occur in 3.9% of all patients with ACS. In-hospital mortality rates were significantly higher in patients with major bleeding than in those without major bleeding (18.6% versus 5.1%, p<0.001), regardless of clinical presentation, and major bleeding was independently associated with an increased risk of hospital death by multivariate analysis.

A subsequent analysis of bleeding events from the Canadian Acute Coronary Registries identified almost 6,000 patients with NSTE ACS who developed in-hospital major bleeding.6 These patients not only had significantly higher rates of in-hospital mortality, MI and stroke, but one-year outcomes of death and MI were also higher in these individuals. On multivariate analysis, major bleeding proved to be a powerful predictor of in-hospital and one-year mortality, MI and death or MI.

Using pooled data from four contemporary ACS trials, Rao et al.7 found that 27.6% of all study participants with available bleeding data had at least one bleeding event during their index hospitalisation. These investigators divided bleeding into mild, moderate and severe categories and found that survival decreased significantly as bleeding severity increased. Additionally, after adjusting for potential confounders, there appeared to be a stepwise increase in the hazard for 30-day mortality, 30-day mortality or MI, and six-month mortality based upon bleeding severity. Interestingly, even patients who developed 'mild' bleeding were found to have significant increases in the adjusted hazard for death and death or MI, suggesting any degree of bleeding in the setting of ACS may have negative prognostic implications.

More recently, Eikelboom et al.9 reviewed bleeding events from the Organization to Assess Ischemic Syndromes (OASIS) Registry,23-25 OASIS-226 and the Clopidogrel in Unstable Angina (CURE)18 trial. In an analysis of 34,126 patients with ACS, they found that major bleeding was associated with a five-fold increase in the risk of death. Similar to previous studies,7 there appeared to be a dose relationship between the severity of bleeding and death, with increasing bleeding severity associated with an increasing risk of death. Furthermore, major bleeding was associated with an increased risk of recurrent ischaemic events, including MI and stroke, even after adjustment for baseline characteristics and propensity for major bleed.

In another study by Rao and colleagues,8 the relative prognostic value of the GUSTO and TIMI bleeding classifications was assessed in 15,858 patients with NSTE ACS. These investigators found that both bleeding scales identified patients at increased risk for adverse outcomes. However, after adjusting for blood transfusion, the stepwise increase in risk with worsening GUSTO bleeding persisted, while the risk with worsening TIMI bleeding was no longer statistically significant. When bleeding definitions were included in the same risk model, only GUSTO bleeding was associated with worse prognosis. Furthermore, when exploring interaction terms between these two scales, it was found that TIMI bleeding did not affect risk among patients who already met criteria for GUSTO bleeding; the converse, however, was not true, suggesting that perhaps a clinically-based assessment of bleeding may be more important for prognosis than a laboratory-guided approach.

Pharmacological Strategies to Reduce Bleeding

Given the prognostic implications of bleeding suggested by these previous studies, more recent clinical trials of pharmacologic agents for the treatment of ACS have made a more concerted effort to both evaluate and target bleeding complications. OASIS-5 randomised over 20,000 NSTE ACS patients to fondaparinux (2.5 mg/day), a direct thrombin inhibitor, or enoxaparin (1.0 mg/kg twice daily).27 The primary end-point of death, MI or refractory ischaemia at day nine was similar between the two study groups, meeting the pre-specified criteria for non-inferiority; however, the rates of major bleed, minor bleed and any bleed were significantly lower in the fondaparinux-treated patients. The finding of increased guiding catheter thrombosis in those receiving fondaparinux in both OASIS-5 and more recently in OASIS-6,28 a study of fondaparinux in patients with STEMI, has raised concern for the routine use of this novel agent.

The evaluation of bivalirudin, another direct thrombin inhibitor thought to have lower bleeding potential, has been studied in patients undergoing percutaneous coronary intervention (PCI). The Randomized Evaluation in PCI Linking Angiomax to Reduced Clinical Events (REPLACE)-2 trial was a randomised, double-blind, active-controlled trial conducted among over 6,000 patients undergoing urgent or elective PCI.18 Patients in this study were randomly assigned to receive intravenous bivalirudin, with provisional glycoprotein IIb/IIIa inhibition, or heparin with planned glycoprotein IIb/IIIa inhibition. The bivalirudin arm of this study was found to be statistically 'not inferior' to the heparin plus planned glycoprotein IIb/IIIa blockade strategy with regard to suppression of acute ischaemic end-points, and was indeed associated with fewer bleeding events.

A retrospective review of the REPLACE-2 data, looking specifically at ACS patients, found a similar rate of death, MI or urgent revascularisation at 30 days compared with a stable cohort of patients, but a higher rate of revascularisation at six months.29 Additionally, a trend for lower major bleeding in the bivalirudin-treated patients was also observed. Results from a more definitive, prospective study of bivalirudin in moderate- to high-risk ACS patients should help to define the role for this pharmacologic agent in future clinical practice.30

Anaemia as a Cardiovascular Risk Factor

In and of itself, anaemia is a relatively common finding in patients with ACS, and may be a marker for chronic illness or the result of bleeding complications from anticoagulant therapy, invasive procedures or both. Already determined to be an independent risk factor for adverse cardiovascular outcomes in community cohorts31 and in patients undergoing PCI,32 a recent review of nearly 40,000 ACS clinical trial participants found a similar, highly significant and independent association between low haemoglobin concentration and a variety of adverse cardiovascular events.33 Individuals with anaemia were more likely to be older, female, and to have worse renal function compared with ACS patients without anaemia. Additionally, those patients with low haemoglobin concentrations were less likely to undergo early revascularisation. The pathophysiology underlying the association between anaemia and adverse cardiovascular outcomes is largely speculative, but likely involves mismatch between myocardial oxygen supply and demand. It is also plausible that changes in physician practice, such as utilising a less invasive management strategy or withholding evidence-based therapeutics that have greater bleeding potential, may also contribute to this clinical association.

Transfusion Therapy in Patients with ACS

Because anaemia and bleeding events are often seen in patients with ACS, blood transfusion therapy is commonly employed. Clinical studies suggest that approximately 10-15% of patients underwent transfusion during their index hospitalisation.4,10 Transfusion therapy was more common in older patients, women, those with a lower body mass index (BMI) and those with multiple medical co-morbidities, with renal insufficiency being most predictive.4,10 In the critical care literature, appropriate use of transfusion therapy remains controversial.34 More recent analyses of transfusion practice in patients with cardiovascular disease will likely fuel this controversy.

Studies of clinical outcomes in cardiac patients receiving blood transfusions have shown disparate results. A trial comparing a restrictive transfusion strategy with a more liberal strategy of transfusion to maintain a haemoglobin level of 10 mg/dl in critically ill patients found no clinical benefit to the more aggressive application of transfusion therapy.35 A post hoc analysis of this randomised trial, limited to patients with cardiovascular disease, further supported those findings.9 On the other hand, a community-based study of elderly patients with acute MI found that blood transfusion had a beneficial effect on survival in individuals with a baseline haematocrit level of 30% or less.36

In ACS trials, however, transfusion therapy seems to be associated with worse clinical outcomes. Pooled data from Platelet IIb/IIIa Antagonism for the Reduction of Acute Coronary Syndrome Events in a Global Organization Network (PARAGON) B, GUSTO IIb, and Platelet Glycoprotein IIb/IIIa in Unstable Angina: Receptor Suppression Using Integrilin Therapy (PURSUIT) showed a higher hazard ratio for death and the composite of death and MI with transfusion therapy in ACS patients. Furthermore, analysis of this data looking specifically at the association between transfusion and nadir haematocrit suggested that a haematocrit level as low as 25% may be tolerated without transfusion therapy in an otherwise stable patient with ischaemic heart disease.10

Recently corroborating these findings, an adjusted analysis of the CRUSADE data revealed that patients who received blood transfusion had longer hospital stays, along with higher absolute rates of death and death or MI.4

Conclusions

Despite advances in invasive and non-invasive therapeutics, which improve outcomes in ACS, bleeding and transfusion remain potential risks. Studies have consistently shown that advanced age, female gender, lower body mass and renal insufficiency are associated with an increased risk for both. Though the exact mechanism of harm has not been clearly delineated, bleeding and blood transfusion are associated with worse clinical outcomes among patients with ACS. Future studies in this patient population will need to address appropriate dosing strategies for pharmacologic agents to reduce the risk of bleeding complications, while still maintaining an effective anticoagulant effect. Additionally, randomised trials of transfusion strategies are needed to determine the appropriate use of blood transfusion in patients with ischaemic heart disease.

References

  1. Braunwald E, Antman EM, Beasely JW, et al, ACC/AHA 2002 guideline update for the management of patients with unstable angina and non-ST-segment elevation myocardial infarction: summary article , Circulation (2002);106: pp. 1893-1900.
    Crossref | PubMed
  2. Bertrand ME, Simoons ML, Fox KA, et al., Management of acute coronary syndromes: acute coronary syndromes without persistent ST segment elevation: recommendations of the Task Force of the European Society of Cardiology , Eur Heart J (2000);21: pp. 1406-1432.
    Crossref | PubMed
  3. Cannon CP, Weintrab WS, Demopoulos LA, et al., Comparison of early invasive versus early conservative strategies in patients with unstable angina and non-ST elevation myocardial infarction treated with early glycoprotein IIb/IIIa inhibition , N Engl J Med (2001);344: pp. 1879-1887.
    Crossref | PubMed
  4. Fox KA, Poole-Wilson P, Clayton TC, et al., 5-year outcome of an interventional strategy in non-ST-elevation acute coronary syndrome: the British Heart Foundation RITA 3 randomised trial , Lancet (2005);366: pp. 914-920.
    Crossref | PubMed
  5. Moscucci M, Fox KA, Cannon CP, et al., Predictors of major bleeding in acute coronary syndromes: the Global Registry of Acute Coronary Events (GRACE) , Eur Heart J (2003);24: pp. 1815-1823.
    Crossref | PubMed
  6. Segev A, Strauss BH, Tan M, et al., Predictors and 1-year outcome of major bleeding in patients with non-ST-elevation acute coronary syndromes: insights from the Canadian Acute Coronary Syndrome Registries , Am Heart J (2005);150: pp.690-694.
    Crossref | PubMed
  7. Rao SV, O'Grady K, Pieper KS, et al., Impact of bleeding severity on clinical outcomes among patients with acute coronary syndromes , Am J Cardiol (2005);96: pp. 1200-1206.
    Crossref | PubMed
  8. Rao SV, O'Grady K, Pieper KS, et al., A comparison of the clinical impact of bleeding measured by two different classifications among patients with acute coronary syndromes , J Am Coll Cardiol (2006);47: pp. 809-816.
    Crossref | PubMed
  9. Eikelboom JW, Mehta SR, Anand SS, et al., Adverse impact of bleeding on prognosis in patients with acute coronary syndromes , Circulation (2006);114: pp. 774-782.
    Crossref | PubMed
  10. Hebert PC, Yetisir E, Martin C et al., Is a low transfusion threshold safe in critically ill patients with cardiovascular disease? , Crit Care Med (2001);29: pp. 227-234.
    Crossref | PubMed
  11. Rao SV, Jollis JG, Harrington RA, et al., Relationship of blood transfusion and clinical outcomes in patients with acute coronary syndromes , J Am Med Assoc (2004);292: pp. 1555-1562.
    Crossref | PubMed
  12. Yang X, Alexander KP, Chen AY, et al., The implications of blood transfusions for patients with non-ST-segment elevation acute coronary syndromes. Results from the CRUSADE National Quality Improvement Initiative , J Am Coll Cardiol (2005);46: pp. 1490-1495.
    Crossref | PubMed
  13. Alexander KP, Chen AY, Roe MT, et al., Excess dosing of antiplatelet and antithrombin agents in the treatment of non- ST-segment elevation acute coronary syndromes , J Am Med Assoc (2005);294: pp. 3108-3116.
    Crossref | PubMed
  14. Huynh T, Piazza N, DiBattiste PM, et al., Analysis of bleeding complications associated with glycoprotein IIb/IIIa receptor blockade in patients with high-risk acute coronary syndromes: insights from the PRISM-PLUS study , Int J Cardiol (2005);100: pp. 73-78.
    Crossref | PubMed
  15. Lim W, Dentali F, Eikelboom JW, Crowther MA, Meta-analysis: low-molecular-weight heparin and bleeding in patients with severe renal insufficiency , Ann Intern Med (2006);144: pp. 673-686.
    Crossref | PubMed
  16. GUSTO Investigators, An international randomized trial comparing four thrombolytic strategies for acute myocardial infarction , N Engl J Med (1993);329: pp. 673-682.
    Crossref | PubMed
  17. Chesebro JH, Knatterud G, Roberts R, et al., Thrombolysis in Myocardial Infarction (TIMI) Trial, I: a comparison between intravenous tissue plasminogen activator and intravenous streptokinase: clinical findings through hospital discharge , Circulation (1987);76: pp. 142-154.
    Crossref | PubMed
  18. Yusuf S, Zhao F, Mehta SR, et al., for the Clopidogrel in Unstable Angina to Prevent Recurrent Events (CURE) 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: pp. 494-502.
    Crossref | PubMed
  19. Lincoff AM, Bittle JA, Harrington RA, et al., Bivalirudin and provisional glycoprotein IIb/IIIa blockade compared with heparin and planned glycoprotein IIb/IIIa blockade during percutaneous coronary intervention: REPLACE-2 randomized trial , J Am Med Assoc (2003);289: pp. 853-863.
    Crossref | PubMed
  20. Francis RC, Hohne C, Kaczmarczyk G, Boemke W, Effect of angiotensin II and endothelin-1 receptor blockade on the haemodynamic and hormonal changes after acute blood loss and after retransfusion in conscious dogs , Acta Physiol Scand (2004);180: pp. 319-328.
    Crossref | PubMed
  21. Ponchon P, Elghozi JL, Contribution of humoral systems to the recovery of blood pressure following severe haemorrhage , J Auton Pharmacol (1997);17: pp. 319-329.
    Crossref | PubMed
  22. Mulvihill NT, Foley JB, Murphy RT, et al., Risk stratification in unstable angina and non-Q wave myocardial infarction using soluble cell adhesion molecules , Heart (2001);85: pp. 523-527.
    Crossref | PubMed
  23. Yusuf S, Flather M, Pogue J, et al., Variations between countries in invasive cardiac procedures and outcomes in patients with suspected unstable angina or myocardial infarction without initial ST elevation: OASIS (Organization to Assess Strategies for Ischemic Syndromes) registry investigators , Lancet (1998);352: pp. 507-514.
    Crossref | PubMed
  24. Chinese Coordinating Center of OASIS Registry, The clinical characteristics of acute coronary syndromes in China , Zhonghua Nei Ke Za Zhi (2003);42: pp. 697-700.
    PubMed
  25. Prabhakaran D, Yusuf S, Mehta S, et al., Two-year outcomes in patients admitted with non-ST elevation acute coronary syndrome: results of the OASIS Registry 1 and 2 , Indian Heart J (2005);57: pp. 217-225.
    PubMed
  26. Organization to Assess Strategies for Ischemic Syndromes (OASIS-2) Investigators, Effects of recombinant hirudin (lepirudin) compared with heparin on death, myocardial infarction, refractory angina, and revascularization procedures in patients with acute myocardial ischemia without ST elevation: a randomized trial , Lancet (1999);353: pp. 429-438.
    Crossref | PubMed
  27. Yusuf S, Mehta SR, Chrolavicius S et al., Comparison of fondaparinux and enoxaparin in acute coronary syndromes , N Engl J Med (2006);354: pp. 1464-1476.
    Crossref | PubMed
  28. Yusuf S, Mehta SR, Chrolavicius S, et al., Effects of fondaparinux on mortality and reinfarction in patients with acute STsegment elevation myocardial infarction: the OASIS-6 randomized trial , J Am Med Assoc (2006);295: pp. 1519-1530.
    Crossref | PubMed
  29. Rajagopal V, Lincoff AM, Cohen DJ, et al., Outcomes of patients with acute coronary syndromes who are treated with bivalirudin during percutaneous coronary intervention: an analysis from the Randomized Evaluation in PCI Linking Angiomax to Reduced Clinical Events (REPLACE-2) trial , Am Heart J (2006);152: pp. 149-154.
    Crossref | PubMed
  30. Stone GW, Bertrand M, Colombo A, et al., Acute Catheterization and Urgent Intervention Triage strategY (ACUITY) trial: study design and rationale , Am Heart J (2004);148: pp. 764-775.
    Crossref | PubMed
  31. Sarnak MJ, Tighiouart H, Manjunath G, et al., Anemia as a risk factor for cardiovascular disease in the Atherosclerosis Risk in Communities (ARIC) study , J Am Coll Cardiol (2002);40: pp. 27-33.
    Crossref | PubMed
  32. McKechnie RS, Smith D, Montoye C, et al., Prognostic implication of anemia on in-hospital outcomes after percutaneous coronary intervention , Circulation (2004);110: pp. 271-277.
    Crossref | PubMed
  33. Sabatine MS, Morrow DA, Giugliano RP, et al., Association of hemoglobin levels with clinical outcomes in acute coronary syndromes , Circulation (2005);111: pp. 2042-2049.
    Crossref | PubMed
  34. Vincent JL, Piagnerelli M, Transfusion in the intensive care unit , Crit Care Med (2006);34(5 Suppl): pp. S96-S101.
    Crossref | PubMed
  35. Hebert PC, Wells G, Blajchman MA et al., Transfusion Requirements in Critical Care Investigators, Canadian Critical Care Trial Groups. A multicenter, randomized, controlled clinical trial of transfusion requirements in critical care , N Engl J Med (1999);340: pp. 409-417.
    Crossref | PubMed
  36. Wu WC, Rathore SS, Wang Y, Radford MJ, Krumholz HM, Blood transfusion in elderly patients with acute myocardial infarction , N Engl J Med (2001);345: pp. 1230-1236.
    Crossref | PubMed
  37. Global Organization Network (PARAGON)-B Investigators, Randomized, placebo-controlled trial of titrated intravenous lamifiban for acute coronary syndromes , Circulation (2002);105: pp. 316-321.
    Crossref | PubMed