Your browser doesn't support JavaScript. Please upgrade to a modern browser or enable JavaScript in your existing browser.
Skip Navigation U.S. Department of Health and Human Services www.hhs.gov
Agency for Healthcare Research Quality www.ahrq.gov
www.ahrq.gov

Chapter 25. Beta-blockers and Reduction of Perioperative Cardiac Events

Andrew D. Auerbach, M.D., M.P.H.
University of California, San Francisco School of Medicine

Background

As the most common complications of major noncardiac surgery, myocardial infarction and cardiovascular death have long been a focus of preoperative evaluations1-4 and a target of perioperative management strategies. Until recently, methods to reduce the incidence of these complications depended upon preoperative assessments of risk combining clinical evaluation with clinical prediction rules, followed by additional tests or revascularization procedures, as appropriate.1 The benefit of preoperative revascularization remains unclear, as no randomized prospective trial has demonstrated its benefit.5 Indeed, concern exists that preoperative intervention might prove detrimental, as the net benefit in terms of reduced perioperative cardiac events may be offset by the risks of the revascularization strategy itself. Newer strategies, including the use of percutaneous transluminal angioplasty as the revascularization modality, may have promise.6 Large prospective trials examining these approaches are underway.5

Strong evidence links myocardial ischemia with postoperative myocardial events.7,8 One study found postoperative ischemia increased the odds of postoperative myocardial events 21-fold.9 Based on findings from observational studies that beta-blockade blunts electrocardiographic signs of ischemia,10-12 recent trials have examined the effects of perioperative beta-blocker administration on patient outcomes. Results of these investigations are extremely promising, and beta-blockade may represent an important new method of reducing perioperative cardiac risk. This chapter reviews the evidence from randomized controlled trials examining the effect of perioperative beta-blockade on cardiac events (i.e., myocardial ischemia, angina, myocardial infarction, pulmonary edema, and cardiac death).

Practice Description

Although published studies have employed different agents, doses, and dosing schedules, the general approach in each study has been similar: administration of a therapeutic dose of beta-blocker prior to induction of anesthesia, followed by beta-blockade through the operation and in the postoperative period. In all regimens, the dose is titrated to a target heart rate, generally 70 beats per minute or lower.

Prevalence and Severity of the Target Safety Problem

Myocardial cardiac events are the most common medical complication of surgery, occurring in 2-5% of patients undergoing non-cardiac surgery13 and as many as 30% of patients undergoing vascular surgery.14,15 Perioperative cardiac events are associated with a mortality rate of nearly 60% per event,14,16 prolonged hospitalization, and higher costs.16,17 The prevalence of these events and their high mortality have made the prevention of perioperative cardiac ischemia the subject of practice guidelines1,17 and numerous prediction rules13,16,18 to detect patients at high risk for these complications.

Opportunities for Impact

As a relatively new therapy, few data describe the use of perioperative beta-blockade in clinical practice. However, evidence suggests it is utilized infrequently. A recent observational study in the Netherlands of patients undergoing vascular surgery showed that only 27% of these high-risk patients received beta-blockers perioperatively.19

Study Designs

Using a structured MEDLINE® search, we identified 4 relevant randomized controlled trials of the effectiveness of perioperative beta-blockade in reducing perioperative cardiac events, including myocardial ischemia and cardiac or all-cause mortality (Table 25.1). A randomized trial by Harwood et al was excluded because both groups received beta-blockers (i.e., there was no control group).20 Although data from a study by Wallace et al21 were derived from one of the randomized trials included in this review,22 it reported effects of beta-blockade upon different outcomes (i.e., myocardial ischemia) and was included in our review. There was sufficient evidence available to limit the review to studies of Level 1 design. Observational studies, such as those by Pasternack et al and Boersma et al,11,19 are not included.

Study Outcomes

The studies identified included a range of clinical outcomes: 2 included assessment of myocardial ischemia12,23 and 3 reported myocardial infarction, pulmonary edema, cardiac death, or all-cause mortality (Level 1 outcomes).15,22,23

Evidence for Effectiveness of the Practice

Of studies reporting the effect of beta-blockers on perioperative ischemia (Level 2 outcome), all but one found a statistically significant reduction in ischemia among treated patients. Wallace et al,21 in a subset analysis of data from Mangano et al,24 reported less frequent perioperative myocardial ischemia in atenolol-treated patients. Stone et al25 suggested a similar effect of beta-blockade on Holter-monitor documented myocardial ischemia. However, the authors did not report the types of procedures included in their sample, nor did they statistically compare baseline patient characteristics, leaving their conclusions open to debate. Raby et al12 also found a significant beneficial effect of beta-blockade using a continuous infusion of esmolol in high-risk patients undergoing vascular surgery. Although Urban et al also found a reduction in perioperative ischemia, this difference failed to reach statistical significance.23 These findings may be explained in part by a relatively low cardiac risk in Urban's cohort, who were undergoing elective total knee replacement. The patients in many of the other studies were at higher risk of cardiac events, as demonstrated by rates of ischemia in the control groups. In studies finding a statistical difference, rates of ischemia were between 28% and 73% in controls, as compared with the 15% rate of ischemia observed in Urban's control group.

Of studies reporting cardiac events and cardiac mortality, 2 reported significant improvement in patient outcomes due to beta-blockade. In a study of male veterans undergoing major noncardiac surgery, Mangano et al22 reported a relative reduction in all-cause mortality of nearly 55% at 2 years. This difference, which appeared within the first 8 months of follow-up, was ascribed to a marked reduction in cardiac events in the first year of therapy (67% reduction at year 1, 48% at year 2). However, patients in the beta-blocker group had less coronary disease at study entry, were on ACE-inhibitors more frequently, and were less likely to have beta-blockers discontinued perioperatively, perhaps biasing results in favor of the treatment group.26, 27 Accounting for these differences in multivariate models of varying stringency did not invalidate their findings.24 Although questions remain about the generalizability of results to other patient populations, the authors favored broader use of beta-blockade in the setting of clinical trials.

Poldermans et al15 suggested an even greater benefit of beta-blockade among high-risk patients. These investigators enrolled patients undergoing vascular surgery who had myocardial ischemia documented by dobutamine echocardiography, with an estimated rate of perioperative cardiac event of 28%. The entire patient cohort experienced a 90% reduction in cardiac death or non-fatal myocardial infarction at 30 days. Follow-up care did not include additional therapy (i.e., cardiac catheterization, revascularization), raising concerns that the research algorithm did not reflect optimal clinical practice.28, 29 However, if the true rate of events in treated patients is low (the point estimate from this small study was 3.4%), the risks associated with revascularization30 may outweigh any benefit.

In contrast to the previous 2 studies, Urban et al23 found no statistically significant difference in rates of in-hospital myocardial infarction. It is likely that these investigators' ability to detect a difference was limited in part by the relatively small sample size and shorter length of follow-up. Other studies of perioperative beta-blockade employed longer periods of follow-up to detect events up to 2 years following surgery.

Differences in absolute magnitude of benefit can be ascribed in part to the cardiac risks of the patients enrolled (again reflected in event rates in the control groups in each study), with the most powerful benefits seen in patients at highest risk. The greater benefit seen in Poldermans et al's study15 may also be due to the fact that the study did not enroll patients who were receiving beta-blockers. Patients who are beta-blocker naïve may have a different response to perioperative use of bisoprolol, or the preexisting use of these agents may represent a confounding factor not completely accounted for in other studies of perioperative beta-blockade.

Beta-blockade may have additional beneficial effects for elderly patients. Patients who received beta-blockers were extubated more quickly, required less medication for pain, and were alert sooner after surgery.31 Although the unblinded nature of this study leaves its findings open to debate, the possibility of additional benefits is tantalizing and worthy of further investigation.

Potential for Harm

Stone et al reported high rates of bradycardia (21/89 patients) in beta-blocker treated patients, "half" of whom required atropine therapy. However, the vague descriptions and more general problems with the study's design make it difficult to interpret the significance of these events in clinical practice. Adverse events related to the use of beta-blockers in other reviewed studies were infrequent (10% or less in Mangano et al22) and did not require therapy or result in withdrawal of the medication. Similar rates of side effects have been noted in studies examining beta-blockade in patients undergoing cardiac surgery.20,32,33 One study examining use of propranolol in patients undergoing thoracotomy for pneumonectomy suggested that patients receiving beta-blockers had twice the rate of postoperative congestive heart failure (4/50 vs. 8/50, p<0.01). In addition, 16% (8/50) of patients in the treatment arm had the drug discontinued due to "bronchospasm."34

Finally, a recent prospective observational study has suggested that withdrawal of beta-blockade from patients immediately following surgery may result in adverse events.35 This effect was not observed in randomized trials of beta-blockade that employed shorter treatment regimens12,25 and should be confirmed by larger studies.

Costs and Implementation

The costs of beta-blockers are generally low, and the systems required to use them according to the protocols used in these studies are already in place. In addition, there is the potential for significant cost-savings if routine use of beta-blockers allows a safe reduction in the use of extensive preoperative cardiovascular testing.

Comment

Results from several well-designed clinical trials suggest that use of beta-blockers in the perioperative period is associated with significant reductions in patient cardiac morbidity and mortality. In the future such therapy may reduce the need for additional tests and revascularization procedures,14 further reducing costs of care. However, several questions regarding its use remain, and should be topics of future research.

First, no clear data suggest an advantage of one particular beta-blocking agent over another. Studies to date have employed several different beta-blockers, suggesting that the efficacy of beta-blockade is class dependent if titrated to physiologically active dosages. Other (alpha-1 selective) sympatholytics also improve patient outcomes,36 raising the possibility that combined alpha-beta antagonists (i.e., labetolol) may have benefit. Second, results from Shammash et al document the hazards of discontinuation of beta-blockers immediately postoperatively,35 and most protocols employed treatment regimens that extended longer—even up to one month following surgery. The current studies suggest beta-blockade should be continued for at least one week postoperatively. Third, effectiveness of beta-blockade in patients at high risk due to aortic stenosis or unstable or severe cardiovascular symptoms (New York Heart Association Class III-IV) is unknown, as these patients were not included in the reviewed studies. Similarly, its utility—both in terms of cardiac events and cost—in patients with very low risk of perioperative cardiac events (i.e., those undergoing same-day or outpatient surgery, ophthalmic surgery, or those who have minimal cardiac risk) is unclear. Beta-blockade has not been studied in patients undergoing regional anesthesia or conscious sedation. In addition, no study to date has examined the use of beta-blockade in patients who have poor functional status and might otherwise be referred for additional non-invasive testing.1,5,14,17

Finally, the increasing popularity of perioperative beta-blockade, particularly catalyzed by the results of the study by Poldermans et al,15 calls into question whether risk stratification using published guidelines or risk indices is still necessary.28 Although beta-blockade is likely to be effective in many patients, the identification of patients at highest risk is still important, as these patients may require additional testing and therapy. A recent study of beta-blockade noted improved outcomes across a spectrum of predicted cardiac risk, but noted that cardiac events could be further reduced in high-risk patients through use of additional non-invasive testing and subsequent "usual care."19 Thus, although beta-blockade may increase the threshold at which clinicians refer patients for additional testing, the era of risk stratification is not over.

The use of beta-blockers to reduce perioperative cardiac events and mortality represents a major advance in perioperative medicine for some patients at intermediate and high risk for cardiac events during noncardiac surgery. Wider use of this therapy should be promoted and studied, with future research focused on fine-tuning dosages and schedules and identifying populations of patients in which its use is cost-effective.

Table 25.1. Randomized controlled trials of the effectiveness of perioperative beta-blockade*

Study Participants Regimen Resultsa Side Effects Comments
Mangano, 199622
Wallace, 199821
200 patients undergoing elective noncardiac surgery Atenolol 5-10g IV 30 min before entry into OR, after surgery, and 50-100g qd through hospital stay (up to 7 days); Target HR 55-65 bpm; doses held if HR<55 bpm or SBP<100 mmHg or defined adverse event

All-cause mortality at 2 yrs: 9% vs. 21% (p=0.019)

Cardiac death at 2 yrs: 4% vs. 12% (p=0.033)

Postoperative ischemia: 24% vs. 39% (p=0.03)

Intraoperative bradycardia more common with atenolol (38% vs. 15%, p=0.0002) but no difference in need for treatment

No increase in third-degree heart block, hypotension, bronchospasm, or congestive heart failure

Included patients already taking beta-blockers, an excess of which (18 vs. 8%) were in the beta-blocker group

NNT 9.1 (primary endpoint)

Poldermans, 199915 112 patients with positive results on dobutamine echocardiography undergoing elective abdominal aortic or infrainguinal arterial reconstruction Bisoprolol 5-10 mg po qd, begun an average of 37 days preoperatively and continued for 30 days postoperatively. Doses held if HR<50 bpm or SBP<100 mmHg

Cardiac death: 3.4% vs. 17% (p=0.02)

Nonfatal MI: 0% vs. 17% (p<0.001)

No exacerbations of peripheral vascular disease

Excluded patients already on beta-blockers

NNT 3.2 (cardiac death or nonfatal MI)

Raby, 199912 26 patients with preoperative ischemia by Holter monitor undergoing aortic aneurysm repair, infrainguinal arterial bypass, or carotid endarterectomy Esmolol IV for 48 hr postoperatively. Titrate to HR 20% below ischemic threshold but no less than 60 bpm Postoperative ischemia: 33% vs. 73% (p<0.05) No patient had beta-blocker therapy suspended because of unacceptable side effects

Clinicians prescribed alternate postoperative beta-blockers more often in control group (13% vs. 82%, p<0.05)

NNT 2.5 (primary endpoint)

Stone, 198837 128 untreated hypertensive patients undergoing elective surgery. Hypertension defined as systolic blood pressure 160-200 mmHg, diastolic 90-100 mmHg Patients randomized to control, labetolol 100 mg po, atenolol 50 mg po, or oxprenolol 20 mg po given before induction of anesthesia Myocardial ischemia: 2/89 (2%) vs. 11/39 (28%) in untreated patients (p<0.001)

21 patients with beta-blockers had bradycardia, "half required atropine."

No bradycardia in control patients

Patients had generally similar baseline characteristics, but these were not statistically compared

No description of surgeries performed

Urban, 200023 120 patients undergoing elective total knee arthroplasty Esmolol IV within 1 hr after surgery, titrate to HR<80 bpm. Change to metoprolol morning of 1st postoperative day. Titrate to HR<80 bpm for next 48 hrs then continue dose until discharge

Postoperative ischemia: 6% vs. 15% (p=NS)

Postoperative MI 2% vs. 6% (p=NS)

None noted Included patients already on beta- blockers (30% in each arm)

* HR indicates heart rate; MI, myocardial infarction; NNT, number needed to treat; and NS, not statistically significant.

a Results are reported as beta-blocker group vs. control group.

References

1. Guidelines for assessing and managing the perioperative risk from coronary artery disease associated with major noncardiac surgery. American College of Physicians. Ann Intern Med 1997;127:309-312.

2. Belzberg H, Rivkind AI. Preoperative cardiac preparation. Chest 1999;115:82S-95S.

3. Merli GJ, Weitz HH. The medical consultant. Med Clin North Am 1987;71:353-355.

4. Merli GJ, Weitz HH. Approaching the surgical patient. Role of the medical consultant. Clin Chest Med 1993;14:205-210.

5. Goldman L. Assessing and reducing cardiac risks of noncardiac surgery. Am J Med 2001;110:320-323.

6. Hassan SA, Hlatky MA, Boothroyd DB, et al. Outcomes of noncardiac surgery after coronary bypass surgery or coronary angioplasty in the Bypass Angioplasty Revascularization Investigation (BARI). Am J Med 2001;110:260-266.

7. Reich DL, Bodian CA, Krol M, Kuroda M, Osinski T, Thys DM. Intraoperative hemodynamic predictors of mortality, stroke, and myocardial infarction after coronary artery bypass surgery. Anesth Analg 1999;89:814-822.

8. Hewer I, Drew B, Karp K, Stotts N. The utilization of automated ST segment analysis in the determination of myocardial ischemia. Aana J 1997;65:351-356.

9. Landesberg G, Luria MH, Cotev S, et al. Importance of long-duration postoperative ST-segment depression in cardiac morbidity after vascular surgery. Lancet 1993;341:715-719.

10. Smulyan H, Weinberg SE, Howanitz PJ. Continuous propranolol infusion following abdominal surgery. JAMA 1982;247:2539-2542.

11. Pasternack PF, Grossi EA, Baumann FG, et al. Beta blockade to decrease silent myocardial ischemia during peripheral vascular surgery. Am J Surg 1989;158:113-116.

12. Raby KE, Brull SJ, Timimi F, et al. The effect of heart rate control on myocardial ischemia among high-risk patients after vascular surgery. Anesth Analg 1999;88:477-482.

13. Lee TH, Marcantonio ER, Mangione CM, et al. Derivation and prospective validation of a simple index for prediction of cardiac risk of major noncardiac surgery. Circulation 1999;100:1043-1049.

14. Lee TH. Reducing cardiac risk in noncardiac surgery. N Engl J Med 1999;341:1838-1840.

15. Poldermans D, Boersma E, Bax JJ, et al. The effect of bisoprolol on perioperative mortality and myocardial infarction in high-risk patients undergoing vascular surgery. Dutch Echocardiographic Cardiac Risk Evaluation Applying Stress Echocardiography Study Group. N Engl J Med 1999;341:1789-1794.

16. Goldman L. Multifactorial index of cardiac risk in noncardiac surgery: ten-year status report. J Cardiothorac Anesth 1987;1:237-244.

17. Eagle KA, Brundage BH, Chaitman BR, et al. Guidelines for perioperative cardiovascular evaluation for noncardiac surgery: an abridged version of the report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Mayo Clin Proc 1997;72:524-531.

18. Detsky AS, Abrams HB, Forbath N, Scott JG, Hilliard JR. Cardiac assessment for patients undergoing noncardiac surgery. A multifactorial clinical risk index. Arch Intern Med 1986;146:2131-2134.

19. Boersma E, Poldermans D, Bax JJ, et al. Predictors of cardiac events after major vascular surgery: Role of clinical characteristics, dobutamine echocardiography, and beta-blocker therapy. JAMA 2001;285:1865-1873.

20. Harwood TN, Butterworth J, Prielipp RC, et al. The safety and effectiveness of esmolol in the perioperative period in patients undergoing abdominal aortic surgery. J Cardiothorac Vasc Anesth 1999;13:555-561.

21. Wallace A, Layug B, Tateo I, et al. Prophylactic atenolol reduces postoperative myocardial ischemia. McSPI Research Group. Anesthesiology 1998;88:7-17.

22. Mangano DT, Layug EL, Wallace A, Tateo I. Effect of atenolol on mortality and cardiovascular morbidity after noncardiac surgery. Multicenter Study of Perioperative Ischemia Research Group. N Engl J Med 1996;335:1713-1720.

23. Urban MK, Markowitz SM, Gordon MA, Urquhart BL, Kligfield P. Postoperative prophylactic administration of beta-adrenergic blockers in patients at risk for myocardial ischemia. Anesth Analg 2000;90:1257-1261.

24. Mangano DT. Effect of atenolol on mortality and cardiovascular morbidity after noncardiac surgery. N Engl J Med 1997;336:1452.

25. Stone JG, Foex P, Sear JW, Johnson LL, Khambatta HJ, Triner L. Myocardial ischemia in untreated hypertensive patients: effect of a single small oral dose of a beta-adrenergic blocking agent. Anesthesiology 1988;68:495-500.

26. Reis SE, Feldman AH. Effect of atenolol on mortality and cardiovascular morbidity after noncardiac surgery. N Engl J Med 1997;336:1453.

27. Petros JA. Effect of atenolol on mortality and cardiovascular morbidity after noncardiac surgery. N Engl J Med 1997;336:1452.

28. Litwack R, Gilligan D, DeGruttola V. Beta-Blockade for patients undergoing vascular surgery. N Engl J Med 2000;342:1051-1053.

29. Feldman T, Fusman B, McKinsey JF. Beta-Blockade for patients undergoing vascular surgery. N Engl J Med 2000;342:1051-1052.

30. Poldermans D, Boersma E. Beta-Blockade for patients undergoing vascular surgery. N Engl J Med 2000;342:1052-1053.

31. Zaugg M, Tagliente T, Lucchinetti E, et al. Beneficial effects from beta-adrenergic blockade in elderly patients undergoing noncardiac surgery. Anesthesiology 1999;91:1674-1686.

32. Hammon JW, Wood AJ, Prager RL, Wood M, Muirhead J, Bender HW. Perioperative beta blockade with propranolol: reduction in myocardial oxygen demands and incidence of atrial and ventricular arrhythmias. Ann Thorac Surg 1984;38:363-367.

33. Lamb RK, Prabhakar G, Thorpe JA, Smith S, Norton R, Dyde JA. The use of atenolol in the prevention of supraventricular arrhythmias following coronary artery surgery. Eur Heart J 1988;9:32-36.

34. Bayliff CD, Massel DR, Inculet RI, et al. Propranolol for the prevention of postoperative arrhythmias in general thoracic surgery. Ann Thorac Surg 1999;67:182-186.

35. Shammash JB, Trost JC, Gold JM, Berlin JA, Golden MA, Kimmel SE. Perioperative beta-blocker withdrawal and mortality in vascular surgical patients. Am Heart J 2001;141:148-153.

36. Oliver MF, Goldman L, Julian DG, Holme I. Effect of mivazerol on perioperative cardiac complications during non-cardiac surgery in patients with coronary heart disease: the European Mivazerol Trial (EMIT). Anesthesiology 1999;91:951-961.

37. Stone JG, Foex P, Sear JW, Johnson LL, Khambatta HJ, Triner L. Risk of myocardial ischaemia during anaesthesia in treated and untreated hypertensive patients. Br J Anaesth 1988;61:675-679.

Return to Contents
Proceed to Next Chapter

 

AHRQ Advancing Excellence in Health Care