Anesthesia for Cardiac Surgical Procedures


Key Points

  • Common cardiac surgical procedures in adults include coronary revascularization with either cardiopulmonary bypass (CPB) or off pump, as well as cardiac valve repair or replacement for valvular regurgitation or stenosis, surgical management of heart failure (e.g., ventricular assist devices, extracorporeal membrane oxygenation, cardiac transplantation), initial repair of or reoperation for congenital heart disease, surgical ablation of atrial fibrillation, pericardiocentesis or pericardiectomy, and repair of traumatic injuries to the heart or thoracic aorta.

  • Patients undergoing “redo” cardiac surgery (i.e., those who have previously had a median sternotomy) warrant special concern about the possibility of sudden, massive hemorrhage. At least 2 units of blood should be immediately available for all such cases.

  • The choice of anesthetic drugs and techniques for inducing anesthesia should include consideration of the patient’s cardiac pathophysiology and other comorbid conditions. Hypotension may result from vasodilation secondary to decreased sympathetic tone induced by anesthetics, particularly in patients with poor left ventricular function. Conversely, hypertension may occur from preinduction anxiety or sympathetic stimulation caused by laryngoscopy and endotracheal intubation.

  • During the prebypass period, hemodynamic and metabolic stability of the patient should be achieved while making preparations for CPB. The degree of surgical stimulation varies markedly during this period.

  • CPB activates the extrinsic and intrinsic coagulation pathways and alters platelet function through the influence of hemodilution, hypothermia, and contact activation by bypass circuit materials.

  • Problems that may occur during the postbypass or postoperative time periods include hypotension resulting from surgical or technical failure, left or right ventricular dysfunction, vasoplegia syndrome, or left ventricular outflow tract obstruction. Other potential problems include dysrhythmias, pulmonary complications (e.g., atelectasis, bronchospasm, mucus or blood plugging of the endotracheal tube, pulmonary edema, hemothorax, pneumothorax), metabolic disturbances (e.g., hypokalemia, hyperkalemia, hypocalcemia, hypomagnesemia, hyperglycemia), and bleeding and coagulopathy.

  • The Society of Thoracic Surgeons (STS), the Society of Cardiovascular Anesthesiologists, and the American Society of ExtraCorporeal Technology published a joint statement on practice guidelines for transfusion and blood conservation in cardiac surgery. A European Guideline published by the European Association for Cardio-Thoracic Surgery and European Association of Cardiothoracic Anesthesiology confirm these recommendations. The guidelines include recommendations regarding the following: (1) use of drugs that decrease postoperative bleeding, including antifibrinolytic drugs; (2) techniques for conserving blood and minimizing hemodilution, including cell saver sequestration, retrograde priming of the pump, and normovolemic hemodilution; and (3) implementation of transfusion algorithms supported with point-of-care testing.

  • The incidence of overt postoperative stroke after isolated coronary artery bypass grafting (CABG) surgery has decreased significantly to 1.2% despite the more frequent and current prevalence of diabetes mellitus and hypertension. The major risk factor for central nervous system injury or dysfunction is particulate and microgaseous emboli; other factors include cerebral hypoperfusion and the inflammatory response to surgery and CPB.

  • STS guidelines state that blood glucose levels should be maintained at less than 180 mg/dL throughout the perioperative period, although many centers treat cardiac surgical patients somewhat more aggressively with continuous insulin infusions in an attempt to keep glucose levels lower than 150 mg/dL. Hypoglycemia should be avoided.

  • Postoperative pain after median sternotomy or thoracotomy causes enhanced sympathetic tone and may lead to myocardial ischemia secondary to increased heart rate, pulmonary vascular resistance, myocardial work, and myocardial oxygen consumption. Furthermore, pain may cause “splinting” that interferes with the patient’s ability to cough and clear secretions and leads to postoperative respiratory insufficiency.

  • Common procedures performed in “hybrid” operating rooms or cardiac interventional suites include electrophysiology procedures and procedures that involve percutaneous management of structural heart lesions. These include valvular lesions and atrial or ventricular septal defects. Other percutaneous procedures performed in non-OR locations include placement of ventricular assist devices, extracorporeal membrane oxygenation, and aortic endovascular stenting.

Acknowledgment

This chapter consolidates the 8th edition chapter of the same title and Chapter 104 Nitric Oxide and Other Inhaled Pulmonary Vasodilators. The returning authors Muhammad F. Sarwar Bruce E. Searles, Linda Shore-Lesserson, and Marc E. Stone as well as the editors and publisher would like to thank the following authors: Nancy A. Nussmeier, Isobel Russell, Fumito Ichinose, and Warren M. Zapol for their contributions to the prior edition of this work. It has served as the foundation for the current chapter.

Cardiovascular Disease in the 21st Century

Age, Gender, and Race

An estimated 82,600,000 U.S. adults (<1 in 3) have one or more types of cardiovascular disease (CVD), of whom approximately half are 60 years old or older. The prevalence of CVD will likely increase because of the “graying” of the United States (i.e., the aging of our population) and the increasing incidences of obesity and hypertension. Although mortality from coronary artery disease (CAD) specifically has decreased since the 1970s, CVD remains the leading cause of death for both men and women in the United States ( Fig. 54.1 ). Furthermore, approximately 7,588,000 inpatient cardiovascular operations and procedures are performed yearly in the United States, with direct and indirect costs totaling approximately $315.4 billion. As the current healthcare reforms expand treatment coverage, these costs will probably increase.

Fig. 54.1, Cardiovascular disease (CVD) and other major causes of death for all male and female members of the U.S. population in 2008.

Despite the common perception that CVD affects mainly men, this is true only in younger age groups. The sex distribution of CVD changes with age; CVD becomes equally prevalent between the sexes by the age of 60 years, and by the age of 80 years, more women than men are affected. The impact of CVD on the health status of U.S. women has gained recognition and is the focus of public education efforts such as the “Go Red for Women” campaign sponsored by the American Heart Association (AHA) and the “Red Dress” project sponsored by the Department of Health and Human Services, the National Institutes of Health, and the National Heart, Lung, and Blood Institute. Furthermore, a series of editorials and articles in the Journal of Thoracic and Cardiovascular Surgery and gender-specific practice guidelines for coronary artery surgery in the Annals of Thoracic Surgery have emphasized sex differences that affect patients who undergo cardiac surgical procedures. For example, using internal mammary artery (IMA) grafts significantly reduces mortality in patients of both sexes, but, until more recently, these grafts were underused in women. Although some studies have shown that women’s short-term survival is worse than men’s after coronary artery bypass grafting (CABG) surgery, others have found that 5-year post-CABG survival is actually better in women than in men.

Mortality resulting from CVD has been consistently more frequent in black than white patients. In 2008, the overall CVD-related death rate per 100,000 persons was 390.4 for black male patients, 287.2 for white male patients, 277.4 for black female patients, and 200.5 for white female patients. Furthermore, racial disparities have been reported in outcomes after CABG: both unadjusted mortality rates and mortality rates adjusted for patient-related characteristics are higher in black patients than in white patients. In fact, as an unfortunate consequence of the public release of quality information through CABG “report cards” in certain states, institutions and individual surgeons may use “racial profiling” in selecting patients for CABG. For example, in New York State, physicians’ avoidance of racial and ethnic minorities, who may be at a higher risk for poor outcomes, may have temporarily widened the disparity in CABG use among white versus black and Hispanic patients.

Genetic Influences in Cardiac Disease

Evolving rapidly beyond racial considerations (which are increasingly losing their medical and scientific meaning in the “melting pot” of the United States), perioperative genomics is the study of the unique biologic makeup of surgical patients. This field holds promise for uncovering the biologic reasons that patients with similar risk factors can have dramatically different perioperative clinical outcomes. Patients with complex comorbid conditions are exposed to controlled trauma in the cardiac surgical operating room ( Fig. 54.2 ). The hope is that preoperative risk assessment and outcome prediction will soon include testing for genetic markers related to individual differences in inflammatory, thrombotic, and vascular responses to stress related to cardiopulmonary bypass (CPB) and to the operation itself.

Fig. 54.2, Biologic systems and mechanistic pathways genetically associated with perioperative adverse events.

One example is the prevention of perioperative myocardial infarction (MI). Mechanisms in the pathogenesis of myonecrosis include the complex acute inflammatory response to surgery and CPB. Functional genetic variants in cytokine and leukocyte-endothelial interaction pathways are independently associated with the severity of myonecrosis after cardiac surgery. Increased concentrations of the most extensively studied inflammatory marker, C-reactive protein (CRP), are associated with increased mortality in patients who undergo CABG. Both increased baseline plasma CRP levels and increased acute-phase postoperative plasma CRP levels are genetically determined. Another pathophysiologic process in perioperative MI is coagulation variability with a tendency toward thrombosis. Polymorphisms in platelet activation and thrombin formation have been associated with myocardial injury and with mortality after cardiac surgery.

Genetic factors have been associated with other postoperative complications. Common genetic variants of CRP and interleukin-6 (IL-6) are significantly associated with the risk of stroke and cognitive decline after cardiac surgery. Angiotensin-converting enzyme (ACE) gene polymorphisms predict a patient’s risk of respiratory complications that necessitate prolonged mechanical ventilation after cardiac surgery.

Research in the field of genetic and molecular determinants of outcomes in patients undergoing cardiac surgery continues. In addition to preoperative risk assessments, intraoperative diagnoses and therefore planning for the postoperative care of the patient will be influenced as new discoveries are made.

Approach to Anesthesia for the Adult Cardiac Patient

Preoperative Evaluation, Preparation, and Monitoring

Heart and Vascular System

Electrocardiography

Standard monitoring of cardiac surgical patients by electrocardiography (ECG) involves using the five-lead electrode system. An electrode is placed on each extremity, and one precordial electrode is placed in the V 5 position (on the left anterior axillary line at the fifth intercostal space). Ischemia detection is greatest (75%) with the V 5 lead. This sensitivity increases to 80% when lead II is paired with a V 5 lead. The addition of a second precordial lead, V 4 , increases the sensitivity, thus making it possible to detect nearly 100% of ischemic episodes. Currently, most ECG monitors can perform ST-segment analysis automatically with high sensitivity and specificity for detecting ischemia.

Despite appropriate lead selection and the use of automated ST-segment monitoring, perioperative ECG monitoring has important limitations. Visual inspection of the ECG on the monitor has low sensitivity in diagnosing ischemic changes, and automated ST-segment analysis depends on the computer’s ability to set the isoelectric and J-point reference points accurately. During cardiac surgical procedures, the set points should be checked before and after bypass, especially persistent changes in heart rate, because the reference points chosen at the beginning of the case may not be accurate under conditions that may arise later.

Arterial and Central Venous Pressure Monitoring (see also Chapter 36 )

Invasive arterial cannulation and monitoring comprise a standard of care for cardiac surgical patients. Patients’ comorbidities often include labile hypertension, atherosclerotic CVD, or both. Furthermore, cardiac surgical technique frequently causes sudden, rapid changes in arterial blood pressure resulting from factors such as direct compression of the heart, impaired venous return caused by retraction and cannulation of the great vessels, and arrhythmias resulting from mechanical stimulation. In addition, sudden, significant blood loss may induce hypovolemia and hypotension. Finally, during nonpulsatile CPB, noninvasive blood pressure recordings are not accurate. Intraarterial monitoring provides continuous, real-time, beat-to-beat assessment of arterial perfusion pressure and waveform throughout the cardiac surgical procedure. Having an indwelling arterial catheter also enables the frequent drawing of arterial blood for laboratory analyses.

Although the radial artery is the most commonly accessed site, the femoral, brachial, ulnar, dorsalis pedis, posterior tibial, and axillary arteries can also be used. The pressures measured in the peripheral arteries are different from the central aortic pressure because the arterial waveform becomes progressively more distorted as the signal is transmitted down the arterial system. Although the mean arterial pressure (MAP) measured in the peripheral arteries is usually similar to the central aortic pressure, this may change after CPB is initiated. When cannulating a radial artery, one should consider the native state of the collateral circulation in the hand and the possible surgical use of a radial artery free graft as an arterial conduit. If a radial arterial graft is to be harvested, it is usually taken from the nondominant hand or arm; thus, the arterial line should be placed on the dominant side.

Central venous access is also standard of care during cardiac surgery. In addition to monitoring central venous pressure (CVP), central venous access provides a portal for intravascular volume replacement, pharmacologic therapy, and the insertion of other invasive monitors, such as pulmonary artery (PA) catheters. In addition, a CVP catheter can be used to measure the filling pressure of the right ventricle and estimate intravascular volume status. Although the CVP does not directly reflect left-sided heart filling pressure, it may be used as an estimate of left-sided pressures in patients with good left ventricular (LV) function. Following trends is more useful than relying on an individual measurement. In many patients, using a central venous catheter may offer a better risk-to-benefit ratio than using a PA catheter. For the accurate measurement of pressure, the distal end of the catheter must lie within one of the large intrathoracic veins or the right atrium.

An internal jugular vein is most commonly selected for catheterization because it provides ease of approach and optimal distance from the operative field. A femoral or subclavian vein can be considered as well, but groin access can be challenging in obese patients and may be inappropriate if femoral bypass cannula placement or vein graft harvesting is necessary. The subclavian approach is also imperfect because it is associated with an increased risk of catheter obstruction during sternal retraction.

The use of ultrasound is being rapidly adopted throughout the United States for facilitating the placement of central venous catheters and reducing the complications associated with them. Although ultrasound-guided central catheter placement is easily accomplished and appears to improve patients’ outcomes, concerns regarding the associated costs of the required hardware and training are reasons for the lack of universal adoption of this technique ( Box 54.1 ).

Box 54.1
Some of the Recognized Benefits of and Concerns About Ultrasound-Guided Central Venous Cannulation
Modified from Kaplan JA, Reich DL, Savino JS, eds. Kaplan’s Cardiac Anesthesia: The Echo Era. 6th ed. St. Louis: Saunders; 2011:426.

Benefits

Ultrasonic guidance of internal jugular vein cannulation can be particularly advantageous in patients with difficult neck anatomy (e.g., short neck, obesity) or prior neck surgery, anticoagulated patients, and infants.

  • Greater success rate on first attempt

  • Fewer overall attempts

  • Better access in patients with difficult neck anatomy (e.g., resulting from obesity or prior surgery)

  • Fewer complications (e.g., carotid artery puncture, anticoagulant-enhanced bleeding)

  • Visible vessel patency, anatomic variants

  • Relatively inexpensive technology

Concerns

  • Need for personnel to be trained to maintain aseptic technique when using sterile probe sheaths

  • Requirement for additional training

  • Inability to show surface anatomy

  • Potential loss of landmark-guided skills when needed for emergency central venous catheterization

Pulmonary Artery Catheterization

The PA catheter is a flow-directed catheter typically placed through an introducer in the central venous compartment via the internal jugular, subclavian, or femoral vein (see also Chapter 36 ). This catheter can measure PA pressure (PAP), CVP, and pulmonary capillary wedge pressure (PCWP). However, the PCWP can both overestimate and underestimate LV filling pressure ( Box 54.2 ). Some PA catheters are designed with a thermistor to register blood temperature changes, which are used to calculate right-sided heart cardiac output (CO) or ejection fraction (EF) by thermodilution. PA catheters may also have oximetric capabilities to measure mixed venous oxygen saturation (
S v ¯ O 2
). Thus, a PA catheter can be used to assess intravascular volume status, measure CO, measure
S v ¯ O 2
, and derive hemodynamic parameters.

Box 54.2
Conditions Resulting in Discrepancies Between Pulmonary Capillary Wedge Pressure and Left Ventricular End-Diastolic Pressure
Modified from Tuman KJ, Carroll CC, Ivankovich AD. Pitfalls in interpretation of pulmonary artery catheter data. Cardiothorac Vasc Anesth Update. 1991;2:1.

PCWP > LVEDP

  • Positive-pressure ventilation

  • PEEP

  • Increased intrathoracic pressure

  • Non-West lung zone III PAC placement

  • Chronic obstructive pulmonary disease

  • Increased pulmonary vascular resistance

  • Left atrial myxoma

  • Mitral valve disease (e.g., stenosis, regurgitation)

PCWP < LVEDP

  • Noncompliant left ventricle (e.g., ischemia, hypertrophy)

  • Aortic regurgitation (premature closure of the mitral valve)

  • LVEDP > 25 mm Hg

LVEDP, Left ventricular end-diastolic pressure; PAC, pulmonary artery catheter; PCWP, pulmonary capillary wedge pressure; PEEP, positive end-expiratory pressure.

The CO, the amount of blood delivered to the tissues by the heart, is of particular interest to cardiac anesthesiologists. The product of stroke volume and heart rate, CO is affected by preload, afterload, heart rate, and contractility. PA catheters capable of measuring CO continuously were introduced into clinical practice in the 1990s. The correlation of continuous CO measurements with those measurements obtained by using the intermittent thermodilution method is good in physiologically and thermally stable prebypass and postbypass periods.

Continuous monitoring of
S v ¯ O 2
provides a means to estimate the adequacy of oxygen delivery relative to oxygen consumption. Decreases in
S v ¯ O 2
may indicate decreased CO, increased oxygen consumption, decreased arterial oxygen saturation, or decreased hemoglobin concentration. If it is assumed that oxygen consumption and arterial oxygen content are constant, changes in
S v ¯ O 2
should reflect changes in CO. However, London and colleagues found that continuous
S v ¯ O 2
monitoring did not lead to better outcomes than standard PA catheter monitoring.

Pacing PA catheters are also commercially available. Electrode PA catheters include five electrodes for atrial, ventricular, or atrioventricular (AV) sequential pacing. Paceport PA catheters (Edwards Lifesciences, Irvine, CA) have a port for the insertion of a ventricular wire or of both atrial and ventricular wires for temporary pacing.

The risk-to-benefit ratio involved in using PA catheters has been a subject of controversy since the 1990s. Complications of PA catheter placement include those mentioned in the section on CVP placement, as well as transient arrhythmias, complete heart block, pulmonary infarction, endobronchial hemorrhage, thrombus formation, catheter knotting and entrapment, valvular damage, and thrombocytopenia. In addition, a common complication is incorrect interpretation of the data obtained from the PA catheter, with resultant incorrect treatment of the patient. Schwann and colleagues published a large, international, prospective observational study showing that using a PA catheter was associated with a more frequent risk of a composite mortality and morbidity outcome than using a CVP alone in patients undergoing CABG surgery. Smaller observational trials have also associated the PA catheter with increased morbidity and decreased survival in cardiac surgical patients.

Currently, the trend in the United States is to be selective in deciding which patients may benefit from a PA catheter, especially with the widespread use of transesophageal echocardiography (TEE). Absolute contraindications to PA catheter placement include tricuspid or pulmonic valvular stenosis, right atrial (RA) or right ventricular (RV) masses, and tetralogy of Fallot. Relative contraindications include severe arrhythmias and newly inserted pacemaker wires (which may be dislodged by the catheter during insertion). Clearly, patients undergoing low-risk cardiac surgical procedures can be managed safely without PA catheter placement. However, many cardiac surgeons and anesthesiologists still use the device in high-risk cardiac operations and in patients with right-sided heart failure (HF) or pulmonary hypertension, particularly to assist in postoperative management ( Box 54.3 ).

Box 54.3
Possible Clinical Indications for Pulmonary Artery Catheter Monitoring
Modified from Kaplan JA, Reich DL, Savino JS, eds. Kaplan’s Cardiac Anesthesia: The Echo Era. 6th ed. St. Louis: Saunders; 2011:435.

Major procedures involving large fluid shifts or blood loss in patients with:

  • Right-sided heart failure, pulmonary hypertension

  • Severe left-sided heart failure not responsive to therapy

  • Cardiogenic or septic shock or multiple organ failure

  • Hemodynamic instability requiring inotropes or intraaortic balloon counterpulsation

  • Surgery of the aorta requiring suprarenal cross-clamping

  • Hepatic transplantation

  • Orthotopic heart transplantation

Transesophageal Echocardiography

TEE is used in many, if not most, cardiac surgical procedures in the current era. See Chapter 37 for a thorough discussion of this extraordinarily valuable diagnostic and monitoring modality.

Central Nervous System

The incidence of overt postoperative stroke after isolated CABG decreased significantly from 1.6% in the year 2000 to 1.2% in the year 2009, despite the greater prevalence of diabetes mellitus and hypertension in the current era. A more subtle entity, postoperative cognitive decline (POCD), or more recently termed delayed neurocognitive recovery as part of the postoperative neurocognitive disorders (PND), has also been described in numerous studies. However, the measurement of POCD is complicated by variations in the tests used to assess cognitive function, the timing of these tests, and the diagnostic and statistical definitions of decline. Furthermore, studies have shown that compared with nonsurgical and healthy controls, patients who undergo CABG have similar rates of cognitive decline 1 year postoperatively.

Risk factors for central nervous system (CNS) injury or dysfunction after cardiac surgery are listed in Box 54.4 . The most common cause is thought to be particulate or microgaseous emboli. Other factors include cerebral hypoperfusion, particularly in patients with cerebrovascular disease, and the inflammatory response to surgery and CPB.

Box 54.4
Mechanisms of and Contributing Factors to Neurologic Lesions
From Kaplan JA, Reich DL, Savino JS, eds. Kaplan’s Cardiac Anesthesia: The Echo Era. 6th ed. St. Louis: Saunders; 2011:1070.

Embolization

Hypoperfusion

Inflammation

Influencing Factors

  • Aortic atheromatous plaque

  • Cerebrovascular disease

  • Altered cerebral autoregulation

  • Hypotension

  • Intracardiac debris

  • Air

  • Cerebral venous obstruction on bypass

  • Cardiopulmonary bypass circuit surface

  • Reinfusion of unprocessed shed blood

  • Cerebral hyperthermia

  • Hypoxia

Monitoring

Transesophageal and Epiaortic Echocardiography

TEE allows direct visualization of the first segment of the ascending aorta, the middle distal segment of the aortic arch, and a good portion of the descending thoracic aorta. However, the distal segment of the ascending aorta and the proximal midportion of the aortic arch cannot be visualized well because of the interposition of the trachea and bronchi between the TEE probe and these aortic structures. Instead, epiaortic imaging with a handheld, high-frequency probe placed over the ascending aorta or aortic arch can be used to visualize aortic segments that are in the TEE probe’s “blind spot.”

The echocardiographic finding of atheromatous disease of the aorta has been linked to CNS injury in cardiac surgical patients. Atherosclerosis of the ascending aorta is present in 20% to 40% of cardiac surgical patients, and the percentage increases with age. The severity of atheromatous disease of the aorta is a strong predictor of death and stroke after CABG ( Table 54.1 ). Using TEE to guide cannula placement or surgical technique significantly reduced the stroke rate in a series of 500 consecutive patients compared with controls in a statewide database. Avoiding instrumentation of the ascending aorta (the “no touch technique”) in patients with severe aortic atherosclerosis has been advocated. The addition of epiaortic scanning of the ascending aorta increases the sensitivity of intraoperative echocardiography in identifying significant atheromatous disease in this segment of the aorta. Certainly, this combination of techniques is superior to surgical palpation in detecting such disease.

Table 54.1
Grading Aortic Atherosclerosis
Modified from Béïque FA, Joffe D, Tousignant G, Konstadt S. Echocardiography-based assessment and management of atherosclerotic disease of the thoracic aorta. J Cardiothorac Vasc Anesth . 1998;12:206–220.
Aortic Atheromatous Disease Echocardiographic Findings
Grade 1 Normal or mild intimal thickening
Grade 2 Severe intimal thickening
No protruding atheromas
Grade 3 Atheroma protruding <5 mm into the lumen
Grade 4 Atheroma protruding ≥5 mm into the lumen
Grade 5 Atheroma of any size with a mobile component

Cerebral Oximetry

Cerebral oximetry uses near-infrared spectroscopy technology similar to that used in pulse oximeters. Light-emitting electrodes are placed on the patient’s forehead, lateral to the midline, and over both frontal cortices. Because the skull is translucent to infrared light, and because oxygenated and deoxygenated blood have different absorption characteristics when exposed to infrared light of two different wavelengths, regional cerebral oxygen saturation (rSO 2 ) can be computed from the returning signal. The design of the cerebral oximeter enables it to measure change from the patient’s own established baseline and to monitor both frontal lobes simultaneously.

The use of near-infrared spectroscopy has been studied in the perioperative setting; a relative decrease in rSO 2 to less than 80% of the preoperative baseline or to absolute levels less than 50% increased the incidence of adverse postoperative outcomes. These outcomes include POCD, stroke, organ dysfunction, mortality, and length of hospital stay. A physiologically derived treatment algorithm for management of perioperative cerebral oxygen desaturation has been proposed ( Fig. 54.3 ). As noted by Murkin, an important confounder in evaluating the role of monitoring cerebral oximetry is the efficacy of treatment for cerebral desaturation.

Fig. 54.3, Algorithm for the use of brain oximetry.

A baseline rSO 2 that is low even though the patient is breathing supplemental oxygen (absolute value ≤50%) is an independent risk factor for 30-day and 1-year mortality. A baseline rSO 2 could act as a refined marker for preoperative risk stratification and help clinicians identify patients who require more intensive monitoring and care in the postoperative period.

Transcranial Doppler

Transcranial Doppler (TCD) involves the ultrasonic interrogation of blood flow velocity through the middle cerebral or common carotid arteries as an indirect measure of cerebral blood flow. The technology has been used extensively as a research tool. For example, in conjunction with cerebral oximetry, TCD has been used to delineate the limits of cerebral autoregulation during CPB. TCD can also detect cerebral emboli. Contrary to previous information, an association between TCD-detected emboli and POCD is questionable.

A primary limitation of TCD technology has been its inability to discern gaseous from solid emboli. Other general limitations of TCD technology include the following: (1) the quality of information is heavily user dependent; (2) accuracy requires stable and precise probe placement, which can be quite cumbersome; and (3) information is affected by patient-related characteristics, such as skin thickness. These difficulties have limited use of this technology in the perioperative setting.

Electroencephalography and Bispectral Index Monitoring

The electroencephalogram (EEG), recorded from multiple adhesive or screw-in scalp electrodes, represents surface cerebral cortical activity (see also Chapter 10, Chapter 9 ). Awake patients produce a pattern of EEG readings that differs from the pattern produced by patients who are under anesthesia. Establishing a baseline and monitoring the changes from that baseline form the premise of EEG monitoring. Changes in the frequency of EEG signals (slower brain waves) and the reduction of wave amplitude may indicate changes in cortical neuronal function that warrant concern.

Multichannel EEG monitoring is not routinely used in cardiac surgery. However, a resurgence of interest in single- or dual-channel processed EEG monitoring, such as the bispectral index (BIS), has occurred. The BIS has been studied as a means to monitor for intraoperative awareness, to reduce overall anesthetic consumption, and to provide information on cerebral perfusion. However, controversy surrounds the usefulness of BIS monitoring in reducing intraoperative awareness. Similarly, the effectiveness of BIS monitoring in either guiding the safe reduction of anesthetic dosing or contributing to the success of fast tracking has not been demonstrated by randomized clinical trials in cardiac surgical patients.

With respect to cerebral ischemia, sudden EEG changes during cardiac surgery and CPB can be attributable to correctable problems such as superior vena cava (SVC) obstruction or severe decrease in CO. The latest iteration of the BIS monitor incorporates bilateral frontal EEG channels, which may increase its ability to detect unilateral frontal ischemia, especially if anesthesia is stable, if the insult is sudden, extended, or located in the frontal area, and if the preoperative EEG was normal. However, many variables may confuse EEG interpretation during cardiac surgery. These include hypothermia, the pharmacologic suppression of EEG signals, and interference produced by pump mechanics. In addition, the EEG measures only cortical activity, so ischemic or embolic injury that occurs below the level of the cortex may go undetected. Therefore, the EEG and derived indices are neither sensitive nor specific in detecting cerebral ischemia.

Summary

Currently, evidence-based recommendations cannot be made regarding the efficacy of treatment for abnormal values. Although not yet recognized as a clinical standard of care, neuromonitoring will likely continue to be the subject of significant research effort.

Renal System

Acute kidney injury (AKI) after cardiac surgery remains a significant cause of postoperative morbidity, increased cost of care, later development of chronic kidney disease, and short-term as well as long-term mortality. Although the pathogenesis of AKI is multifactorial, control of some specific factors may limit its incidence in cardiac surgical patients. Bellomo and associates identified six major injury pathways of cardiac surgery–associated AKI: toxins (both exogenous and endogenous), metabolic factors, ischemia-reperfusion injury, neurohormonal activation, inflammation, and oxidative stress.

Randomized trials of specific potential preventive measures for AKI after cardiac surgery are few. Certainly, all potentially nephrotoxic drugs should be avoided in the perioperative period ( Box 54.5 ). Hydration is, of course, a universally accepted component of strategies to prevent contrast nephropathy. Unfortunately, no pharmacologic strategy has definitive efficacy in preventing early AKI. Atherosclerosis of the ascending aorta appears to be an independent risk factor for AKI.

Box 54.5
Drugs That Contribute to Kidney Injury
Modified from Bellomo R, Kellum JA, Ronco C. Acute kidney injury. Lancet. 2012;380:756–766.

  • Radiocontrast agents

  • Aminoglycosides

  • Amphotericin

  • Nonsteroidal antiinflammatory drugs

  • β-Lactam antibiotics (specifically contribute to interstitial nephropathy)

  • Sulfonamides

  • Acyclovir

  • Methotrexate

  • Cisplatin

  • Cyclosporine

  • Tacrolimus

  • Angiotensin-converting enzyme inhibitors

  • Angiotensin receptor blockers

Intraoperative TEE should be used to identify patients at increased risk for thromboembolic phenomena. Although observational studies have suggested a possible benefit of off-pump procedures and avoidance of aortic manipulation, definitive evidence is lacking. For cardiac surgical cases that require CPB, the duration of aortic cross-clamping should be limited if possible, especially in patients who are at a higher-than-normal risk for renal complications, such as patients with preexisting renal insufficiency. Hemodynamic instability should be addressed quickly, and intravascular volume should be maintained or rapidly restored. Finally, perioperative hyperglycemia should be avoided.

Clearly, definition of measures that may prevent AKI after cardiac surgery is needed. The cost of this complication to the patient and to society is probably higher than previously thought.

Endocrine System

Glucose Control

Hyperglycemia in surgical patients is a consequence of the inflammatory or stress response to the trauma of surgery. Components of this response include an endocrine response (i.e., increased production of counterregulatory hormones such as cortisol, growth hormone, glucagon, and catecholamines) ( Fig. 54.4 ), an immune response resulting in increased cytokine production, an autonomic response resulting in increased sympathetic stimulation, and altered insulin signaling. These changes increase glucose production, decrease glucose elimination during CPB, and induce insulin resistance, thereby causing hyperglycemia.

Fig. 54.4, (A) Plasma levels of epinephrine (Epi) during cardiac surgery. Bars indicate standard error of the mean. X-clamp, Cross-clamp. (B and C) Levels of cortisol during cardiac surgery. ECC, Extracorporeal circulation; Midop, intraoperatively; N.S., not significant; op, operatively; PCV, packed cell volume; Postop, postoperatively; Preop, preoperatively.

All patients are at risk for developing hyperglycemia during cardiac surgery. Older patients, diabetic patients, and patients with CAD are particularly prone to perioperative hyperglycemia. Although cardiac surgery without CPB initiates a stress response, CPB increases this response manyfold. The degree of hyperglycemia depends on several variables associated with the use of CPB, such as the pump prime fluid selected and the degree of hypothermia induced. Epinephrine and other inotropic drugs may contribute to hyperglycemia after CPB by stimulating hepatic glycogenolysis and gluconeogenesis.

Impaired fasting glucose blood levels before cardiac surgery and persistently increased glucose levels during and immediately after surgical procedures are predictive of longer hospital stay and increased perioperative morbidity and mortality in both diabetic and nondiabetic patients. However, in diabetic patients undergoing cardiac operations, hyperglycemia may only partly explain the increased risk for adverse outcomes. Immunologic abnormalities common to diabetic patients, such as decreased chemotaxis, phagocytosis, opsonization, bacterial killing, and antioxidant defense, also promote adverse outcomes by increasing diabetic patients’ risk of infection.

Glucose blood levels should be controlled, beginning in the preoperative period and continuing until discharge. However, in a classic study, patients were randomly assigned to intensive intraoperative insulin treatment (to maintain glucose levels at 80-100 mg/dL) or to standard insulin treatment (to keep glucose levels <200 mg/dL). Surprisingly, the investigators reported a statistically nonsignificant increase in the incidence of death and stroke in the group receiving intensive insulin treatment. Current Society of Thoracic Surgeons (STS) guidelines state that blood glucose levels should be kept lower than 180 mg/dL throughout the perioperative period. Some cardiac surgical centers more aggressively administer continuous insulin infusions in an attempt to keep glucose levels lower than 150 mg/dL.

Thyroid Hormone

Fig. 54.5 shows the effect of thyroid hormone on cardiovascular hemodynamics. Abnormal thyroid function ultimately affects cardiac function in many ways ( Table 54.2 ). During cardiac surgery, the effect of CPB on thyroid hormone production is uncertain. Both increased and decreased levels of thyroid hormone can occur during or immediately after CPB. Free triiodothyronine (T 3 ), the biologically active form of the thyroid hormone, is frequently reduced in cardiac patients because of the reduced activity of the 5′-monodeiodinase responsible for converting thyroxine (T 4 ) into T 3 in peripheral tissues. Patients with low T3 values are predisposed to low CO and therefore death as a complication of cardiac surgery. Patients should be profiled for T3 levels and labelled high risk if the levels are low preoperatively.

Fig. 54.5, Effects of thyroid hormone on cardiovascular hemodynamics.

Table 54.2
Effects of Thyroid Dysfunction on Hemodynamics and Cardiac Function
Reprinted with permission from Klein I, Ojamaa K. Thyroid hormone and the cardiovascular system. N Engl J Med . 2001;344:501–509. Copyright © 2001 Massachusetts Medical Society.
Parameter Normal Values Hyperthyroidism Hypothyroidism
Blood volume (% of normal value) 100 105.5 84.5
Heart rate (beats/min) 72-84 88-130 60-80
Cardiac output (L/min) 4.0-6.0 >7.0 <4.5
Systemic vascular resistance (dyne•s•cm −5 ) 1500-1700 700-1200 2100-2700
Left ventricular ejection fraction (%) >50 >65 ≤60
Isovolumic relaxation time (ms) 60-80 25-40 >80

Notably, hypothyroidism is more common in female than in male patients undergoing CABG. Zindrou and associates noted a 17% higher mortality rate in women who underwent CABG while receiving thyroxine replacement therapy for hypothyroidism. A review of the literature by Edwards and colleagues concluded that ensuring a perioperative euthyroid state in women with hypothyroidism who were undergoing CABG could be helpful in reducing the perioperative mortality of these patients.

Hematologic System

Bleeding is the primary complication of cardiac operations that require CPB. In fact, 10% to 15% of blood product use in the United States is associated with cardiac surgery, and this percentage is increasing, largely because of the increasing complexity of cardiac surgical procedures. Real-world data obtained from a large sample of patients and entered into the STS Adult Cardiac Surgery Database suggest that 50% of patients who undergo cardiac surgical procedures receive a blood transfusion. Complex cardiac operations such as “redo” procedures, aortic operations, and the implantation of ventricular assist devices (VADs) require blood transfusion much more often than do simpler operations. Donor blood is viewed as a scarce resource that is associated with increased healthcare costs and significant risk to patients. Furthermore, perioperative blood transfusion is associated with worse short-term and long-term outcomes. Hence, reducing bleeding and blood transfusions has become a major focus of quality improvement efforts in cardiac surgery.

Heparin as an Anticoagulant

Since its discovery by Jay McLean, MD, in 1915, heparin has stood the test of time and remains the primary anticoagulant used in cardiac operations that require CPB. The mechanism underlying heparin’s anticoagulant effect centers on the heparin molecule’s ability to bind simultaneously to antithrombin III and thrombin. Modern nomenclature refers to antithrombin III as antithrombin (AT). The binding process is mediated by a unique pentasaccharide sequence that binds to AT. The proximity of AT and thrombin, mediated by the heparin molecule, allows AT to inhibit the procoagulant effect of thrombin by binding to the active-site serine residue of the thrombin molecule. The inhibitory effect of AT is increased 1000-fold in the presence of heparin. The heparin-AT complex can affect several coagulation factors, but factor Xa and thrombin are the most sensitive to inhibition by heparin, and thrombin is 10 times more sensitive to the inhibitory effects of unfractionated heparin than is factor Xa.

Only approximately onethird of the heparin molecules in a dose of heparin contain the critical pentasaccharide segment that is needed for high-affinity binding to AT. Thus, relatively large doses are required to produce the anticoagulant effect necessary for CPB. In fact, dosing of heparin for CPB is somewhat empiric. After a baseline activated clotting time (ACT) is measured (the normal range is 80-120 seconds), a dose of 300 to 400 units/kg of heparin is given as an intravenous bolus. Commercially available assays are used for calculating the patient’s dose-responsiveness to heparin in vitro. Some practitioners administer heparin at the dose that is indicated by such an in vitro dose-response assay. Subsequent heparin dosing for extracorporeal circulation (ECC) is targeted at maintaining ACT values longer than 400 to 480 seconds. Also available is a heparin concentration monitor that uses protamine titration analysis for ex vivo calculation of the whole blood heparin concentration. This result is often used as an adjunct to the ACT value in confirming an adequate heparin concentration for CPB. Unfortunately, ACT test results vary substantially with clinical conditions and the particular platform used for measurement. Thus, the evidence supporting the use of a threshold of 400 or 480 seconds is almost entirely anecdotal.

The dose of heparin used in patients on CPB is based on early landmark work published by Bull and coauthors in 1975. A small study that sought evidence of thrombin activity during CPB in nonhuman primates and pediatric patients found results supporting a safe lower limit for ACT of 400 seconds. In 1979, Doty and colleagues proposed a simplified dosing regimen guided by ACT values without dose-response curves. The data and recommendations from these few studies constitute the primary basis for current heparin dosing protocols.

Despite heparin’s historic and continued role in anticoagulation for patients maintained on CPB, it is not a perfect anticoagulant. Intrinsic and extrinsic pathway coagulation occurs despite heparin administration, and platelets can still be activated by contact with bypass circuitry and by heparin directly. Alternative anticoagulants are discussed briefly in the section on heparin-induced thrombocytopenia (HIT).

Monitoring of Anticoagulation

Using ACT to monitor the effectiveness of heparin is not an exact science. Tremendous variability is observed in patients’ anticoagulation responses to a given dose of heparin; reasons for this variability include variations in levels of heparin-binding proteins and AT. Hence, ACT values correlate poorly with actual heparin concentrations. Nevertheless, since the publication of Bull and colleagues’ early work, ACT has been the mainstay of anticoagulation monitoring in cardiac operations that require CPB.

Many different ACT measurement devices are commercially available, and each uses a different platform for clot detection and endpoint signaling. However, they all involve the addition of whole blood to a tube or channel containing a contact-phase activator. The activator can be celite, kaolin, glass, or any combination thereof. The sample is warmed to 37°C before the measurement technique is performed. Clot formation occurs, and the measurement ends when a change in velocity, pressure, oscillation, electromagnetic forces, or even color is observed, depending on the particular platform of measurement.

Several clinical variables can affect the ACT ( Table 54.3 ). In addition to physiologic variations, the design of ACT measurement devices (which varies across their many manufacturers) also affects ACT normal and therapeutic values. ACTs also correlate poorly with whole blood and plasma heparin levels, and responses to heparin differ somewhat between adult and pediatric patients. Some authors argue that, because of its poor correlation with heparin concentration, ACT alone is not an adequate monitor of heparin efficacy, and that simultaneous or adjunct monitoring of heparin concentration should also be used during CPB. Prolongation of ACT by non–heparin-related clinical factors, such as hypothermia, hemodilution, or quantitative or qualitative platelet abnormalities, is a well-documented phenomenon, and the anesthesiologist must understand these factors to determine whether it is safe to reduce the heparin dose when the ACT is prolonged. The poor correlation between ACT and measured heparin concentration also makes it possible that a dose reduction will render the heparin concentration inadequate, even when the ACT remains within an acceptable range.

Table 54.3
Clinical Variables That Can Affect the Activated Clotting Time
Hemodilution Prolongs ACT in heparinized patients
Hypothermia Prolongs ACT
Thrombocytopenia Prolongs ACT
Platelet inhibitors Prolongs ACT
Platelet lysis Shortens ACT
Aprotinin Prolongs only celite ACT
Surgical stress Shortens ACT
ACT, Activated clotting time.

Some point-of-care (POC) monitors, such as the Hepcon HMS system (Medtronic Perfusion Systems, Minneapolis, MN), use a protamine titration assay to calculate the heparin concentration. Despotis et al. suggest that monitoring and maintaining heparin concentration—and thus giving larger doses of heparin—actually protect the hemostatic system and may decrease transfusion requirements. However, other investigators have not been able to confirm that higher doses of heparin confer better hemostatic protection; markers of ongoing coagulation are essentially the same whether traditional ACT monitoring or heparin concentration monitoring is used. The 2018 STS, Society of Cardiovascular Anesthesiologists (SCA), and the American Society of ExtraCorporeal Technology (AmSECT) Guidelines state: “Use of heparin concentration monitoring in addition to ACT might be considered, for the maintenance of CPB, as this strategy has been associated with a significant reduction in thrombin generation, fibrinolysis, and neutrophil activation. However, its effects on postoperative bleeding and blood transfusion are inconsistent (class IIb, Level of Evidence B).” An additional recommendation includes: “During CPB, routine administration of heparin at fixed intervals, with ACT monitoring, might be considered and offers a safe alternative to heparin concentration monitoring. (class IIb, Level of Evidence C).”

The high-dose thrombin time (HiTT) is a modification of the thrombin time that is designed to measure the high levels of heparin that are used during CPB. Unlike ACT, the HiTT correlates well with heparin concentration, both before and during CPB, and it is not affected by hemodilution and hypothermia. As a measure of thrombin inhibition, the HiTT is a more specific test of heparin’s effect on thrombin than ACT, and it appears to possess less artifactual variability. Preoperative heparin infusions do not affect HiTT values.

Protamine and Reversal of Anticoagulation

Protamine, which has been in clinical use for as long as heparin has, remains the heparin reversal drug of choice in cardiac surgery. The protamine dose required to reverse heparin is somewhat controversial. In the first published study to examine this question, Bull and associates chose a dose of 1.3 mg protamine for every 100 units of heparin to provide a slight excess of protamine from a projected needed amount of 1.2 mg for every 100 units of heparin. Protamine is usually administered according to the total amount of heparin given (i.e., 1-1.3 mg protamine per 100 units of heparin). This method may result in luxuriant protamine doses, which reduce any theoretic or real risks of heparin rebound but may put the patient at higher risk for bleeding events because of the anticoagulant effect of protamine. Guidelines for the practice of anticoagulation in CPB recommend the following:

  • 1.

    “It is reasonable to limit the ratio of protamine/heparin to less than 2.6 mg protamine/100 Units of heparin, since total doses above this ratio inhibit platelet function, prolong ACT, and increase the risk of bleeding (class IIa, Level of Evidence C).”

  • 2.

    Because of the risk of heparin rebound in patients requiring high doses of heparin and with prolonged CPB times, low-dose protamine infusion (25 mg/h) for up to 6 hours after the end of CPB may be considered as part of a multimodality blood conservation program (class IIb, Level of Evidence C).

Protamine can also be administered at a dose calculated from the heparin concentration, which is measured by a protamine titration assay. Guidelines do support this practice stating that “it can be beneficial to calculate the protamine reversal dose based upon a titration to existing heparin in the blood, since this technique has been associated with reduced bleeding and blood transfusion” (class IIa, Level of Evidence B). If the heparin concentration is not measured, the protamine dose can be graphically derived by plotting ACT values throughout the case and creating heparin dose-response curves. The amount of protamine used in this method is based on the circulating concentration of heparin in the patient at the time of reversal. Because, theoretically, no excess protamine exists, these patients may be at risk for heparin rebound and therefore may require additional protamine. In a small study conducted in patients undergoing valve surgery, administering protamine in two divided doses by titration resulted in a larger dose but reduced bleeding, presumably by treating heparin rebound.

Unique Hematologic Considerations in Cardiac Surgery

Hematologic Effects of Cardiopulmonary Bypass

The hematologic effects of CPB are complex. Exposure of blood to the surfaces of the extracorporeal circuit is a profound stimulus for inflammatory system upregulation, and activation of the hemostatic system is a component of the normal inflammatory response. According to traditional models of hemostasis, ECC activates both the intrinsic and extrinsic coagulation pathways and directly impairs platelet function. Intrinsic pathway activation can occur by contact activation and the conversion of factor XII to factor XIIa on the various surfaces of the CPB circuit. The tissue factor generated from the wound and the circulating tissue thromboplastin combine to cause the extrinsic activation of coagulation by cell-mediated hemostasis, which involves tissue factor–bearing leukocytes and activated endothelial cells. Tissue factor pathway generation of thrombin has a primary role in CPB-associated hemostasis abnormalities ( Fig. 54.6 ).

Fig. 54.6, Coagulation can be activated during cardiopulmonary bypass (CPB) by the intrinsic pathway, with surface adsorption and activation of factor XII, high-molecular-weight kininogen (HMWK) , and prekallikrein (PK) . Activation of the extrinsic pathway during CPB occurs with tissue injury, as well as with systemic inflammation, and it leads to monocyte and endothelial expression of tissue factor (TF) . TF, in combination with factor VIIa, starts the common pathway with the activation of factor X to factor Xa. Assembly of the prothrombinase complex on phospholipid surfaces leads to the production of thrombin and conversion of fibrinogen to fibrin. Tissue factor pathway inhibitor (TFPI) inhibits TF/VIIa. Thrombin can overcome this TFPI blockade by activating factors XI, VIII, and V and initiating activation of factor X by the tenase complex. K, Kallikrein.

In addition to activating both extrinsic and intrinsic coagulation pathways, CPB directly impairs platelet function through a variety of mechanisms. Platelets express on their surface numerous glycoproteins that serve as receptors for several circulating ligands, such as fibrinogen, thrombin, and collagen ( Fig. 54.7 ). The components of the bypass circuit adsorb circulating proteins that can serve as foci for platelet attraction and adherence. These surface-bound platelets activate and release the contents of their cytoplasmic granules, which can then serve as localized sources of thrombin generation, or they may embolize to initiate microvascular thrombosis.

Fig. 54.7, Platelet activation by cardiopulmonary bypass materials is facilitated by rapid adhesion and conformational alteration of plasma proteins, including von Willebrand factor (vWF) and fibrinogen (Fib) . Platelets adhere to vWF through the glycoprotein (Gp) Ib/IX/V surface glycoprotein and to fibrinogen through the Gp IIb/IIIa receptor. Platelet adhesion stimulates intracellular signaling, which leads to degranulation of α granules (platelet factor 4 [PF4] and β-thromboglobulin [βTG] ) and phospholipid reorganization with the formation of coagulation complexes and fibrin formation. Release of adenosine diphosphate (ADP) from dense granules and activation of the Gp IIb/IIIa receptor also occur and lead to the aggregation of platelets to the adherent layer. Because of shear forces, adherent and aggregated platelets can detach from the membrane and circulate in a degranulated state or form small microaggregates that lodge in the distal vasculature.

Fibrinolytic activity is also increased by CPB. Contact activation leads to the activation of factor XII, prekallikrein, and high-molecular-weight kininogen, which causes endothelial cells to produce tissue plasmin activator, and lysis of fibrin and fibrinogen ensues ( Fig. 54.8 ).

Fig. 54.8, The fibrinolytic system results in the degradation of fibrin by plasmin.

The vascular endothelium is itself an active substrate that is sensitive to circulating mediators, and it expresses and releases anticoagulant and procoagulant factors. When exposed to hypoxia or inflammatory mediators during CPB, the endothelium responds and can induce a relatively prothrombotic state marked by tissue factor upregulation, accelerated platelet adhesion, and increased expression of leukocyte adhesion molecules ( Fig. 54.9 ).

Fig. 54.9, Procoagulant endothelial cell activation.

Heparin Resistance, Altered Heparin Responsiveness, and Antithrombin

Heparin resistance is marked by the inability to raise the ACT to therapeutic levels after administration of the recommended doses of unfractionated heparin. Some investigators have defined heparin resistance as an ACT of less than the target (400-480 seconds) after 600 to 800 units/kg of intravenous heparin are administered. Others have defined heparin resistance as an ACT of less than 400 seconds at any time during the course of CPB and heparin administration. Heparin resistance can result from a congenital deficiency or abnormality of AT, which requires treatment with AT to restore heparin’s anticoagulant properties. More often, however, heparin resistance is the result of an acquired condition caused by the patient’s disease status and physiologic state. Giving these patients larger doses of heparin may augment the ACT value; therefore, a more accurate term for describing these clinical findings is altered heparin responsiveness. This alteration can result from an acquired AT deficiency, increased levels of heparin-binding proteins, activated platelets, sepsis, or other conditions. In a small study of cardiac surgical patients stratified by their history of preoperative heparin use, altered heparin responsiveness was found in approximately 40% of the patients who had received preoperative heparin therapy.

Reported risk factors for altered heparin responsiveness include AT levels less than 60% of normal, preoperative heparin therapy, and a platelet count greater than 300,000/μL. Ranucci and associates also found that low postoperative AT levels were predictive of a longer length of stay in the ICU, and others have associated low AT levels with adverse myocardial outcomes. Not all heparin resistance is AT mediated; thus, it is critical to understand the physiologic factors that contribute to the altered heparin responsiveness so that appropriate treatment can be instituted.

The most common treatment for altered heparin responsiveness is supplemental heparin. In refractory cases, treatment with AT concentrate, or recombinant AT, in doses that are calculated to produce 80% to 100% AT activity, restores heparin responsiveness. AT supplementation is a class I indication for heparin resistance that is due to AT deficiency. Thus the practice of replacing AT using this specific factor replacement is preferred over the use of plasma transfusion, which is no longer recommended due to the morbidity of allogeneic transfusion. When AT is used for this purpose, careful neutralization with protamine and attention to hemostasis are essential because AT augments the effect of heparin and thereby slightly increases postoperative bleeding.

Heparin Rebound

Heparin rebound is clinical bleeding that occurs within approximately 1 hour of protamine neutralization. It is accompanied by coagulation test results indicating residual heparinization, such as a prolonged partial thromboplastin time (PTT) or thrombin time and increased anti-factor Xa activity. Mechanisms of heparin rebound include slow dissociation of protein-bound heparin after protamine clearance, more rapid clearance of protamine than of heparin, lymphatic return of extracellular sequestered heparin, and the clearance of an unknown heparin antagonist. Heparin rebound is rare, yet it is more likely to occur when the protamine dose is based on the residual heparin concentration at the end of CPB than when protamine is given as a ratio to the total heparin administered, because using this ratio usually results in a slight “overdose.” With coagulation monitoring, heparin rebound is easily prevented or treated with supplemental protamine. A protamine infusion has also been successful in preventing heparin rebound in patients in whom heparin rebound is a risk. These include cases in which high doses of heparin were administered or in whom CPB time was prolonged. Perfusion management guidelines suggest that a low dose protamine infusion (25 mg/h) for up to 6 hours after the end of CPB may be considered as part of a multimodality blood conservation program (class IIb, Level of Evidence C).

Heparin-Induced Thrombocytopenia

HIT is an immune-mediated prothrombotic disorder that occurs in patients exposed to heparin. Antibodies form against the protein platelet factor 4 (PF4) when PF4 has formed a complex with heparin. Although PF4 is found in only trace amounts in human plasma and is stored in platelet α granules, the presence of heparin increases plasma PF4 concentrations 15- to 30-fold by displacing bound PF4 on endothelial cell surfaces. PF4 is also expressed on the surface membrane of activated platelets by membrane fusion with the α-granule membrane; thus, PF4 is exposed and available to bind with heparin. The resulting PF4-heparin complex on the platelet surface is recognized by a specific immunoglobulin G (IgG), which binds to the complex and leads to immunologically mediated platelet activation. Hyperaggregability of these activated platelets is the hallmark of HIT and is responsible for its prothrombotic complications. Often a clinical score such as the 4 Ts score can be used to determine whether a heparin-platelet antibody test should be performed to diagnose HIT (class IIa, Level of Evidence B). Diagnosis can be difficult in the postcardiac surgery patient and often the 4 Ts score is unreliable.

A common presentation in patients with HIT is a reduction in the platelet count to less than 100,000/μL or to less than 50% of the baseline count. The incidence of seroconversion after CPB and heparin exposure is quite frequent (20%-50%). However, the reported prevalence of clinical HIT after CPB is only 1% to 3%. Thus, the risk of HIT in cardiac surgical patients with postoperative seroconversion is less than 10%.

The strength of the immunologic response, not the mere presence of PF4 or heparin antibodies, may determine which patients are prone to HIT and are at risk for thromboembolic complications. The presence of preoperative antibody, in addition to postoperative antibody, has been associated with increased morbidity after cardiac surgery. This morbidity takes the form of gut ischemia, renal dysfunction, limb ischemia, and other prothrombotic events.

Management of the cardiac surgical patient with HIT must include a careful risk-to-benefit analysis. The likelihood that a patient has true disease and is at increased risk for a thrombotic event must be weighed against the risks posed by using an alternative anticoagulant to heparin. The urgency of the surgical procedure is also an important factor that can affect decision making. It is preferable, when possible, to defer the operation until antibody titers have become undetectable or only weakly positive, which may occur after 90 days (class IIa, Level of Evidence C). If surgical postponement is not practical, then other therapeutic options must be considered ( Boxes 54.6 and 54.7 ). Currently, the direct thrombin inhibitors are used as the anticoagulants of choice. Hirudin and argatroban are approved by the U.S. Food and Drug Administration (FDA) for use in patients with HIT-related thrombosis. The use of these drugs as anticoagulants for CPB is fraught with hemorrhagic complications. Bivalirudin has been approved by the FDA for use in percutaneous interventions and, because of its short half-life, has been favored as an anticoagulant for CPB in patients with HIT. Guidelines suggest that in patients with a diagnosis of HIT and in need of an urgent operation requiring CPB, anticoagulation with bivalirudin is a reasonable option (class IIa, Level of Evidence B). However, no drug other than heparin has FDA approval for specific use as an anticoagulant in patients undergoing CPB. Bivalirudin undergoes renal elimination. Therefore, in seropositive HIT patients who have significant renal dysfunction, anticoagulation for urgent operations requiring CPB can be accomplished with argatroban, plasmapheresis prior to heparin to remove antibodies, or heparin with concomitant antiplatelet agents to prevent platelet activation (tirofiban, iloprost) (class IIb, Level of Evidence C). These latter two techniques have risk because they include heparin, and have been fraught with increased risks of bleeding. Boxes 54.6 and 54.7 summarize therapeutic options and alternative anticoagulant strategies for the patient with HIT whose operation cannot be deferred until seronegativity is documented. Fig. 54.10 delineates how each alternative drug inhibits factor Xa, thrombin, or fibrinogen.

Box 54.6
Therapeutic Options for Anticoagulation for Cardiopulmonary Bypass in Patients With Heparin-Induced Thrombocytopenia
Modified from Kaplan JA, Reich DL, Savino JS, eds. Kaplan’s Cardiac Anesthesia: The Echo Era. 6th ed. St. Louis: Saunders; 2011:966.

  • 1.

    Ancrod

  • 2.

    Low-molecular-weight heparin or heparinoid (test first)

  • 3.

    Alternative thrombin inhibitor (hirudin, bivalirudin, argatroban)

  • 4.

    Using a single dose of heparin, promptly neutralizing it with protamine, and

    • a.

      Delaying surgery so antibodies can regress or

    • b.

      Using plasmapheresis to decrease antibody levels or

    • c.

      Inhibiting platelets with iloprost, aspirin and dipyridamole (Persantine), abciximab, or RGD blockers

In all cases:

  • 1.

    No heparin in flush solutions

  • 2.

    No heparin-bonded catheters

  • 3.

    No heparin lock intravenous ports

No agent is currently indicated for anticoagulation in cardiopulmonary bypass.

RGD, Receptor glycoprotein–derived.

Box 54.7
Potential Alternative Anticoagulants for Cardiopulmonary Bypass
From Kaplan JA, Reich DL, Savino JS, eds. Kaplan’s Cardiac Anesthesia: The Echo Era. 6th ed. St. Louis: Saunders; 2011:967.

  • Ancrod

  • Low-molecular-weight heparins

  • Factor Xa inhibitors

  • Bivalirudin or other direct thrombin inhibitors (hirudin, argatroban)

  • Platelet receptor inhibitors

Fig. 54.10, Alternatives to heparin.

Protamine Reactions

Protamine is associated with several hemodynamic effects that can be categorized by their presentation and mechanism. Adverse reactions to protamine range from moderate hypotension to more profound and hemodynamically significant reactions that can increase in-hospital mortality risk. The clinical presentation of these reactions serves as a starting point for understanding their mechanistic relationships. Commonly, these reactions are classified as type I, type II, or type III. A type I protamine reaction involves isolated hypotension, with normal to low filling pressures and normal airway pressures. This reaction is usually mild and responds to volume infusion, slowing of protamine infusion, and the gentle titration of vasoactive medications. Type II reactions include moderate to severe hypotension and features of anaphylactoid reactions, such as bronchoconstriction. Anaphylactoid reactions include protamine sensitivity reactions that are classically immunologic or allergic in that they are immunoglobulin E (IgE) antibody mediated. Nonimmunologic mechanisms may involve IgG antibodies or complement activation. Type III reactions are thought to be caused by large heparin-protamine complexes that lodge in the pulmonary circulation, cause the release of mediators, and result in severe hypotension and elevated PA pressures that may lead to acute RV failure. This is obviously a profound response, resulting in global cardiovascular collapse or necessitating the reinstitution of CPB because of intractable RV failure. Fortunately, catastrophic hypotension and intractable RV failure are relatively rare events on the spectrum of protamine reactions.

Mechanistic explanations for protamine reactions include endothelial nitric oxide release, mast cell degranulation, and histamine release associated with rapid infusion. A study by Kimmel and colleagues found that neutral protamine Hagedorn insulin use, documented fish allergy, and a history of nonprotamine medical allergies were independent risk factors for protamine hypersensitivity reactions. In this study, 39% of patients who presented for cardiac surgery had one or more of these risk factors. Other possible but unconfirmed risk factors include prior exposure to protamine, a history of vasectomy, decreased LV function, and hemodynamic instability. The site of injection does not influence the incidence of protamine reactions. Pretreatment with histamine blockade is not preventive.

The following principles summarize treatment options for patients at risk for protamine reactions:

  • 1.

    Protamine should be administered slowly (i.e., over ≥5 minutes). Limit protamine dose to less than 2.6 mg protamine/100 units of heparin, since total doses above this ratio inhibit platelet function, prolong ACT, and increase the risk of bleeding (class IIa, Level of Evidence C).

  • 2.

    In patients with documented adverse events related to protamine, consideration should be given to not rechallenging the patient with protamine. Pharmacologic alternatives to protamine can be considered, or a decision can be made not to reverse heparin. Consideration may be given to using non–heparin-based CPB, performing off-pump coronary artery bypass (OPCAB) with an alternative to heparin, or, if heparin is used, administering nonprotamine heparin reversal drugs such heparinase, or simply waiting for heparin’s effects to dissipate.

  • 3.

    Hypotension associated with protamine reversal of heparin often is ameliorated by simply slowing or pausing protamine infusion while volume is infused through intravenous lines or the aortic cannula. Vasoactive medications, such as phenylephrine or ephedrine, use of calcium chloride, or increased inotropic support may be necessary.

  • 4.

    Severe or intractable hypotension, with or without evidence of pulmonary circulatory involvement, bronchospasm, or overt RV failure, demands immediate aggressive attention, intervention, and planning for a potential return to CPB. Steps to consider include the following:

    • a.

      Reheparinization to prepare the patient for a return to CPB and to reduce heparin-protamine complex size. If hemodynamics permit, a low dose of heparin (70 units/kg) may be tried first while supportive treatment continues, followed by a full CPB dose of heparin (300 units/kg) if it becomes necessary to return the patient to CPB. (class I, Level of Evidence C).

    • b.

      Inotropic support, either by infusion or by intermittent bolus administration, is warranted. Epinephrine and norepinephrine are acceptable options, and milrinone may be considered if the patient’s hemodynamic status permits.

    • c.

      If the patient’s hemodynamic status allows, nebulized albuterol is helpful in the management of bronchospasm and elevated airway pressures.

Antifibrinolytic Therapy: Prophylaxis for Bleeding

Prophylactic use of antifibrinolytic drugs before CPB reduces bleeding and transfusion requirements in cardiac surgical patients in randomized trials and in multiple metaanalyses. The most well-known antifibrinolytic drugs include the synthetic lysine analogues ε-aminocaproic acid (EACA) and tranexamic acid (TA) and the serine protease inhibitor aprotinin. Presumably, the blood-sparing effects of the synthetic drugs result from the inhibition of fibrinolysis by their binding to the lysine-binding sites on plasmin. This also has platelet protective properties because plasmin’s antiplatelet effects are also inhibited by antagonist binding.

Aprotinin is a direct enzymatic inhibitor of plasmin and has other protease-inhibiting properties that confer its antiinflammatory and antikallikrein effects. However, aprotinin was notably associated with increases in post-CPB creatinine values and other adverse organ system outcomes in large-scale observational trials. When a randomized prospective trial showed an increase in mortality in the aprotinin group, despite a reduction in bleeding, the drug was removed from the global market. Although the causes of death in the aprotinin-treated patients were not found to be related to thrombosis or other drug-related effects, aprotinin was rendered unavailable for commercial use for years after publication of this study. The decision to remove aprotinin from clinical use was revisited on reevaluation of these study data, and aprotinin has been reintroduced in Canada and other countries specifically for use as labeled in CABG surgery.

Hypercoagulable States

The use of antifibrinolytics has become common in cardiac operations that require CPB. The use of antifibrinolytic drugs and prohemostatic drugs and blood products to treat bleeding has brought to light the risk of thrombosis in CPB, during which feedback mechanisms are critical and homeostasis is perturbed. All patients incur this risk of thrombosis as consumptive coagulopathy increases, but the risk is greatly increased in patients with congenital or acquired thrombophilic states. These states may be important in view of current practices that involve pharmacologically inhibiting fibrinolytic pathways in cardiac surgical patients.

The factor V Leiden (FVL) mutation is the most common inherited thrombophilic disorder; its prevalence is 3% to 7% in white populations. Mechanistically and clinically, FVL has been implicated in thrombotic complications in cardiac operations, most often those involving a period of circulatory arrest and the use of antifibrinolytics. In a review of FVL mutation, Donahue gave the following summation and recommendations :

  • Cardiac surgical patients who are heterozygous for the FVL mutation bleed less than do noncarriers.

  • The risk of early graft thrombosis may be increased in patients with FVL deficiency.

  • The use of antifibrinolytic drugs may increase thrombotic risk in patients with FVL.

  • Anecdotal evidence suggests that in patients with FVL mutation who are exposed to deep hypothermic circulatory arrest, antifibrinolytics increase thrombotic risk.

Cardiac Surgical Patients Taking Anticoagulant or Antithrombotic Drugs

Antithrombotic therapy in cardiac surgical patients has many roles and applications. Patients with ischemic heart disease can be managed short- or long-term with pharmacologic drugs that may include aspirin, AT inhibitors (heparins), direct thrombin inhibitors, or an array of platelet inhibitors (adenosine diphosphate [ADP] receptor inhibitors and glycoprotein IIb/IIIa [Gp IIb/IIIa] receptor inhibitors). Patients with a history of peripheral vascular disease, valvular heart disease, or low ventricular ejection states can similarly be managed with some form of antithrombotic therapy that may also include warfarin. Frequently, patients arrive for surgery while receiving multiple antithrombotic medications. Thus, postoperative bleeding is a common but challenging complication of cardiac surgery, especially when the bleeding risk posed by CPB itself is considered.

The use of percutaneous coronary interventions such as angioplasty and intracoronary stent deployment for ischemic heart disease has led to the use of antithrombotic medications to maintain stent patency and to prevent stent thrombosis. The American College of Cardiology and AHA (ACC/AHA) initial guidelines for percutaneous coronary intervention recommended (on the basis of class I evidence) the use of both aspirin and clopidogrel for at least 1 year after drug-eluting stent placement. However, percutaneous coronary intervention data with the second generation drug-eluting stents indicate that shorter periods of dual anti-platelet therapy are equally effective in preventing in-stent thrombosis, and thus allow for earlier cessation of a single anti-platelet agent after 6 months. This opens the door for interventions and surgical procedures to be performed sooner and with less risk of bleeding. The administration of thienopyridine antiplatelet drugs in addition to aspirin increases the risk of bleeding after cardiac surgery ; however, it is unclear whether aspirin alone increases this risk. Abundant evidence (mostly level B evidence from small, retrospective, nonrandomized studies) suggests that the ADP receptor antagonist clopidogrel (Plavix) has been associated with excessive perioperative bleeding in patients who undergo CABG. This trend has even been reported in the OPCAB population, although not consistently. Earlier recommendations included a 5- to 7-day delay after discontinuation of clopidogrel in patients who require CABG. However, guidelines suggest that a 3-day delay may be sufficient to lessen bleeding risk and provide safe outcomes. It is likely that a 5- to 7-day delay is not necessary, but some interval between the discontinuation of clopidogrel and CABG is supported by the available evidence.

Patients with a history of Gp IIb/IIIa receptor blockade as a part of their acute coronary syndrome management are at risk of increased bleeding and blood component use when they are given abciximab, especially within 12 hours of cardiac surgery. Shorter-acting Gp IIb/IIIa receptor antagonists do not seem to increase bleeding or adverse outcomes and in fact may improve myocardial outcomes when Gp IIb/IIIa blockers are in use. The interval between the discontinuation of antiplatelet therapy and cardiac or noncardiac surgery is critical to preventing thrombotic events without increasing the risk of bleeding. These decisions regarding the cessation of therapy can be guided by knowledge of drug pharmacology and testing of antiplatelet drug efficacy.

Enoxaparin, a low-molecular-weight heparin, increases transfusion rates and the risk of surgical reexploration when it is used in association with CPB. Low-molecular-weight heparin may also decrease heparin responsiveness.

Patients who present for cardiac surgery with residual warfarin effects may benefit by having enhanced anticoagulation during CPB. Postoperatively, if excessive bleeding is noted and is confirmed by hemostasis testing, factor replacement can be performed with blood products or pharmacologic prothrombin complex concentrates (PCCs).

Patients with atrial fibrillation may be maintained on new antithrombotic therapeutic drugs, direct oral anticoagulants (DOACs) that include thrombin inhibitors (dabigatran) and factor Xa inhibitors (rivaroxaban, apixaban, and edoxaban). These drugs are potent and long-acting, and they have no antidote, so one would expect that treating cardiac surgical patients with these drugs would increase their bleeding risk. When compared to vitamin K antagonists, the DOACs have similar thromboprophylaxis efficacy yet fewer bleeding complications. An additional benefit is that DOACs have a predictable pharmacodynamic profile and routine monitoring is often unnecessary. Routine monitoring tests such as the INR and aPTT do not even accurately assess anticoagulant activity of the DOACs and thus a thrombin time or a direct measure of anti-Xa activity would be considered more accurate.

In summary, patients who present for cardiac surgery with preexisting, pharmacologically induced inhibition of the hemostatic system may have undesirable post-CPB bleeding. The diagnosis and treatment of this complication should be the same whether the derangement is a function of CPB itself, coexisting pharmacologic inhibition, or both. The management and treatment of persistent postoperative bleeding are discussed in the section on problems in the postoperative period.

Induction of Anesthesia and the Prebypass Period

Premedication

The anesthesiologist should ensure that appropriate pre-medications are administered with a sip of water on the morning of surgery. With a few exceptions, patients should receive their usual long-term medications, particularly β-adrenergic blocking drugs, on the day of surgery. The clinician should be aware that ACE inhibitors (ACEIs), if administered on the day of surgery, may increase the patient’s propensity for hypotension. With respect to aspirin, it is recognized that aspirin administration in the early postoperative period may reduce the risk of ischemic complications after CABG surgery. However, patients who receive aspirin immediately preoperatively may have more mediastinal bleeding and greater transfusion requirements. A consensus statement published by the STS recommends that low-intensity antiplatelet drugs (e.g., aspirin) be discontinued before cardiac surgery to reduce patients’ blood transfusion requirements, but this should be done only in purely elective cases in patients without acute coronary syndromes.

However, drugs that inhibit the platelet P2Y12 receptor should be discontinued before operative coronary revascularization (either on-pump or off-pump), if possible. The interval between drug discontinuation and operation is determined from the drug’s pharmacodynamics but may be as short as 3 days for irreversible inhibitors of the P2Y12 platelet receptor. POC tests are available to measure platelet ADP responsiveness. POC tests that show normal platelet ADP responsiveness after administration of an initial dose of clopidogrel indicate P2Y12 resistance with as much as 85% specificity. This is called “high on-treatment platelet reactivity.” Flow cytometry may be more specific in diagnosing the degree of platelet inhibition, but it cannot be measured at the point of care.

The prospect of undergoing cardiac surgery provokes anxiety in most patients. Furthermore, the insertion of intravenous and arterial catheters is painful and must be done before anesthesia is induced. The resultant anxiety and pain can lead to undesirable sympathetic stimulation, with consequent tachycardia and hypertension. The first step in preventing this cycle is thoroughly explaining the anticipated anesthetic techniques and procedures to the patient. Premedication with a narcotic or anxiolytic drug, or both, to mitigate pain and anxiety is usually indicated before the patient is transported to the operating suite. Supplemental intravenous drugs—commonly midazolam and possibly fentanyl—are usually necessary during radial artery cannulation before anesthesia is induced. However, in patients with low CO secondary to congestive HF (CHF), sedation should be performed judiciously to avoid myocardial depression and resultant hypotension. Moreover, in patients with significant pulmonary hypertension, oversedation and respiratory depression leading to hypercapnia or hypoxia must be avoided.

Induction of Anesthesia

In preparing for induction, the clinician should have the following drugs immediately available: vasopressors (e.g., phenylephrine, ephedrine, calcium chloride, readily available vasopressin), one or more inotropes (e.g., ephedrine; epinephrine; readily available norepinephrine, dopamine, or dobutamine), one or more vasodilators (e.g., nitroglycerin, nitroprusside, nicardipine), an anticholinergic drug (atropine), antiarrhythmic drugs (e.g., lidocaine, esmolol, magnesium, amiodarone, adenosine), and heparin. Commonly administered drugs should be drawn up and ready for administration by bolus or infusion, as appropriate; agents that are used less commonly should be readily available in the operating room. Protamine should be readily available, but many institutions require that protamine be stored in unique packaging or at a separate, nearby location to prevent inadvertent premature administration.

Furthermore, the selected antibiotic should be ready to administer according to Surgical Care Improvement Project guidelines. The STS recommends a cephalosporin as the primary prophylactic antibiotic for cardiac surgery; the drug should be administered within 1 hour before incision. In patients who are allergic to penicillin, vancomycin is administered within 2 hours of incision. Finally, antifibrinolytic drugs are commonly used to minimize bleeding and the need for transfusion during cardiac surgery. The most commonly used antifibrinolytic drugs are the synthetic lysine analogues TA and EACA; both reduce total blood loss and decrease the number of patients who require blood transfusion during cardiac procedures.

Anesthetic drugs and techniques for inducing anesthesia should be selected with consideration of the patient’s cardiac pathophysiology and other comorbid conditions. No single “recipe” can guarantee hemodynamic stability during anesthetic induction. Hypotension may result in a patient who is relatively hypovolemic and receives a vasodilator or whose sympathetic tone is reduced by anesthesia. Hypotension is particularly common in patients with poor LV function. Conversely, in patients with good myocardial function, hypertension may occur during induction because of preinduction anxiety or sympathetic stimulation caused by laryngoscopy and endotracheal intubation.

The radial artery or an alternative site should be cannulated before induction of anesthesia to monitor arterial pressure on a beat-to-beat basis. If the radial artery is being harvested as a vascular conduit, the contralateral radial or brachial artery or a femoral artery can be cannulated. Basic monitors, including the ECG and pulse oximeter, should also be used during the induction of anesthesia. During any cardiac surgical procedure, central venous access is necessary to secure so that volume infusion, transfusion therapy, and vasoactive drug administration can be easily delivered directly to the central circulation. A central venous catheter or a PA catheter can be placed either before or after anesthesia is induced. Placement before anesthesia induction is ideal so that the CVPs can be monitored during the induction of anesthesia. However, the placement of these lines in the awake patient can take more time and create discomfort, thus causing unwanted hypertension and tachycardia. The risk-benefit analysis usually dictates that the central venous line be placed after anesthesia induction. The urinary bladder catheter, nasogastric tube, TEE probe, and any additional temperature monitors (e.g., a nasopharyngeal probe) are positioned after induction of anesthesia.

When choosing anesthetic drugs and doses during induction and maintenance, one should consider any pharmacodynamic properties that could affect arterial blood pressure, heart rate, or CO, as well as the desirability of “early” extubation of the trachea (i.e., within a few hours after the operation is completed). Anesthesia is most commonly induced with an opioid and a sedative-hypnotic (etomidate, thiopental, propofol, or midazolam). All anesthetics decrease arterial blood pressure by decreasing sympathetic tone, decreasing systemic vascular resistance (SVR), inducing bradycardia, or directly depressing myocardial function. The only exception is ketamine, which has sympathomimetic effects; however, in patients with catecholamine depletion, ketamine’s sympathomimetic effects may not counterbalance its direct negative inotropic effects. Because of their pharmacologic complexities, all anesthetic agents should be administered judiciously in patients who are critically ill or who have poor LV function.

Muscle relaxants are usually given early in the sequence of anesthetic induction, particularly if relatively large doses of opioids are administered, to minimize chest wall rigidity (see also Chapter 27). With the routine use of fast-track anesthesia techniques, including a trend toward earlier extubation, volatile anesthetics are often chosen as the primary maintenance anesthetic. The predominant effect of isoflurane, desflurane, and sevoflurane is dose-dependent vasodilation with resultant decreases in SVR and arterial blood pressure. These volatile anesthetics may have an advantage in inducing preconditioning, which is particularly important in patients undergoing either CABG with CPB or OPCAB, in which myocardial ischemic insults are likely. The volatile anesthetic agents have several cardioprotective effects, including triggering the preconditioning cascade and mitigating reperfusion injury. However, nitrous oxide probably should be avoided because it can increase gaseous bubble size and adversely affect pulmonary vascular resistance (PVR).

The Pre - Cardiopulmonary Bypass Period

After anesthesia is induced, several important details must be remembered, especially positioning (see also Chapter 34 ). Methods of positioning the arms vary according to institutional practice, but one must avoid causing brachial plexus injury by hyperextending the arms, ulnar nerve injury by improperly padding the olecranon, radial nerve injury by compressing the upper part of the arm against the sternal retractor support posts, or finger injury by entrapping the finger against the metal edge of the surgical table. Proper positioning also ensures that arterial catheters previously placed in the radial, ulnar, or brachial arteries are not “dampened.” The head should be padded and occasionally repositioned during the procedure to prevent occipital alopecia, which can occur several days postoperatively. The eyes should be taped, possibly lubricated, and definitely free from pressure. Pressure-related injury to any soft tissue will potentially be exacerbated by hypothermia and decreased perfusion during CPB. All monitors and tubing should be checked after final positioning to ensure that none are kinked, entrapped, tangled, or inaccessible. Additionally, antibiotics must be administered (with documentation) within 1 hour of incision (vancomycin within 2 hours). Arterial blood gases and blood chemistry (electrolytes, glucose, and calcium), as well as baseline ACT, should be measured shortly after anesthesia is induced. If a continuous mixed venous PA catheter has been inserted, mixed venous hemoglobin oxygen saturation should be measured to calibrate the device.

During the prebypass period, the anesthesiologist’s main goal is to maintain the hemodynamic and metabolic stability of the patient while making preparations for CPB. The degree of surgical stimulation varies markedly during this period. Positioning the patient, inserting additional monitors, preparing the skin, and harvesting the saphenous vein or veins cause only minimal sympathetic stimulation. Therefore, hypovolemic patients and those with poor ventricular function may be susceptible to hypotension during these periods. In contrast, chest incision, sternal splitting, and harvesting of the IMA involve more intense surgical stimulation. These events may cause hypertension, tachycardia, and dysrhythmias, even in previously hypotensive patients. However, just before CPB is initiated, during the cannulation of the great vessels, surgical stimulation is again minimal, and manipulation of the heart and great vessels may transiently decrease venous return and cause a precipitous decline in blood pressure. The anesthesiologist must be ready to treat all hemodynamic aberrations with the vasopressor, inotropic, vasodilator, antiarrhythmic, and anticholinergic drugs mentioned earlier.

In preparation for CPB, anticoagulation must be achieved. Heparin is still the standard drug used and is administered through a central venous catheter at an initial dose of 300 to 400 units/kg. The onset of anticoagulation is almost immediate, but generally, the drug is allowed to circulate for 3 to 5 minutes before its effect is measured. The ACT must increase to at least 300 seconds before CPB is initiated, although most institutions use at least 400 seconds as their standard. Additional heparin is administered, if necessary, to increase the ACT to the desired level. Subsequently, it is common to administer an antifibrinolytic drug (EACA or TA) in an attempt to minimize bleeding and the need for transfusion during cardiac surgery.

After heparinization, the next major step in the prebypass phase is vascular cannulation. One or more large veins or the right atrium is cannulated so that all systemic venous blood is diverted to the pump oxygenator. Additionally, a large artery, usually the ascending aorta, is cannulated so that oxygenated blood is delivered back to the arterial circulation. Heparin is always administered before cannulation. Usually, arterial cannulation is established before venous cannulation to allow rapid intravascular volume or blood resuscitation if necessary. Complications of aortic cannulation include arterial dissection, hemorrhage and resultant hypotension, inadvertent cannulation of the aortic arch vessels, and embolic phenomena caused by dislodged atherosclerotic plaque or by air introduced into or entrained around the aortic cannula. Complications of venous cannulation include hypotension from blood loss, dysrhythmias, and surgical mechanical compression of the heart or great vessels. When arterial cannulation is successful and the cannula has been inspected to ensure that no air is present, volume can be administered in 100-mL increments to treat bleeding and hypovolemia. If necessary, dysrhythmias are treated by cardioversion, medications, or rapid initiation of CPB.

Patients undergoing redo cardiac surgery (i.e., those who have previously had a median sternotomy) warrant special concern about the possibility of sudden, massive hemorrhage. At least 2 units of blood should be immediately available for all redo cases. Frequently, the surgeon will elect to use an oscillating saw in these patients, but mediastinal structures adherent to the underside of the sternum may nevertheless be injured. If the right atrium, right ventricle, great vessels, or an existing coronary graft is cut, the surgeon may elect to initiate CPB on an emergency basis. Therefore, the anesthesiologist should have a systemic dose of heparin prepared. As soon as the patient is heparinized, the femoral or aortic arterial cannula is inserted, and the cardiotomy suckers can be used to create venous return (so-called sucker bypass).

Onset of Cardiopulmonary Bypass

Preparing for the onset of CPB brings about a new set of challenges for a cardiac anesthesiologist. As a preparation for CPB, the surgeon would start by placing purse string sutures in the ascending aorta for the eventual placement of aortic cannula. It is imperative that at this point the anesthesiologist keeps the patient’s blood pressure in a range that would not jeopardize the integrity of the aorta while the cannula is placed. A systolic blood pressure of 90 to 110 mm Hg is traditionally accepted for this stage of the procedure. Around this time heparin will also be administered. Usually 300 units/kg patient weight is administered for a target ACT of 450 to 500 seconds, to safely start the CPB. After the safe placement of aortic cannula, remaining cannulas will be placed by the surgeon, followed by start of the CPB.

During the abovementioned process, a cardiac anesthesiologist monitors the patient’s hemodynamics and rhythm for any undesired changes.

With the onset of CPB, to guard against malposition of the aortic or venous cannula, the perfusionist checks the aortic inflow line pressure and for signs of inadequate venous return while the anesthesiologist checks for persistently low arterial pressure, unilateral blanching of the face, or any swelling in the neck veins, face, or conjunctiva. Once full bypass is established and aortic ejection by the heart has ceased, ventilation and inhaled drugs can be discontinued. If a PA catheter is present, it is pulled back 3 to 5 cm to minimize the risk of pulmonary perforation as the pulmonary arteries collapse. Prebypass urine output is recorded and emptied so that urine output during CPB can be monitored separately. The TEE probe may be used to watch for LV distention with the onset of CPB, which may indicate aortic valve regurgitation or other hemodynamic problems. Once CPB is established, the probe is left in the unlocked (neutral) position until the cardiac chambers are de-aired and the patient is weaned from CPB.

To ensure adequate anesthetic depth, supplemental intravenous sedative-hypnotics are administered, or a volatile agent is administered through a vaporizer connected to the oxygenator gas inlet of the CPB circuit. Administration of muscle relaxant is continued to prevent spontaneous ventilation, movement, or shivering during hypothermia and rewarming.

Weaning From Cardiopulmonary Bypass

After the completion of surgery on CPB, the patient is prepared for coming off of the CPB and the resumption of patient’s own physiology.

As part of a cardiac anesthesiologist’s responsibilities it is of paramount importance that a plan is made beforehand for this part of cardiac surgery. The plan should take into account the nature of surgery, the length of bypass run, length of the aortic cross clamp and patient’s presurgical cardiac status and comorbidities.

In preparation of weaning the patient off of the CPB several issues need to be addressed before a successful weaning process is started. Issues that need to be addressed include temperature, electrolytes (specifically potassium), rhythm, systemic blood pressure, contractility, and any air in the left ventricle (LV).

After addressing the abovementioned issues, the perfusionist gradually allows more and more blood to be pumped by the heart instead of the bypass machine. During this time the cardiac anesthesiologist ensures that any inotropic and/or volume requirements of the patient are met to successfully bring the CPB run to its conclusion.

The “CVP” Mnemonic

Fortunately, for most patients, separation from CPB is a relatively uneventful process. A review by Licker and colleagues emphasized that the key to successful weaning from bypass is clear communication among members of the operating room team. In a study conducted at the Mayo Clinic in Rochester, Minnesota, a strong correlation was noted between the frequency of technical errors and poor communication or coordination among the surgeon, anesthesiologist, and perfusionist.

Regarding clinical issues, several criteria should be met in all cardiac surgical cases before weaning from CPB is attempted. Morris and colleagues suggest a mnemonic, “CVP,” to help the clinician remember the main tasks necessary for the successful termination of CPB ( Table 54.4 ). Each letter of CVP represents several tasks or important points to remember that begin with that letter.

Table 54.4
Elements of Morris, Romanoff, and Royster s Central Venous Pressure Mnemonic for Weaning Patients from Cardiopulmonary Bypass
From Morris BN, Romanoff ME, Royster RL. The postcardiopulmonary bypass period: weaning to ICU transport. In: Hensley FA, Martin DE, Gravlee GP, eds. A Practical Approach to Cardiac Anesthesia . 4th ed. Philadelphia: Lippincott Williams & Wilkins; 2008:230–260.
C V P
Cold Ventilation Predictors
Conduction Visualization Pressure
Cardiac output Vaporizer Pressors
Cells Volume expanders Pacer
Calcium Potassium
Coagulation Protamine

The first “C” stands for cold, which refers to the patient’s temperature at the time of weaning from CPB, which should be 36°C to 37°C. Neither the temperature of the venous blood returning to the CPB circuit nor the nasopharyngeal temperature should ever exceed 37°C because hyperthermia may increase the risk of postoperative neurologic complications (see the section on “Temperature”).

The second “C” stands for conduction, which refers to cardiac rate and rhythm. A heart rate of 80 to 100 beats/min is usually desirable. Bradycardia is treated with epicardial pacing wires and/or with β-adrenergic drugs that have chronotropic and dromotropic, as well as inotropic, properties. Tachycardia (i.e., heart rate >120 beats/min) is also undesirable. Sinus tachycardia may result from anemia, hypovolemia, “light” anesthesia, or the administration of chronotropic drugs; treatment is tailored to the presumed cause. Rhythm is also an important factor in optimizing CO. Third-degree AV block requires pacing, ideally AV pacing. Sinus rhythm is preferable, particularly in patients with poor LV compliance, who are especially dependent on an “atrial kick” to achieve adequate filling. If supraventricular tachycardia is present, direct synchronized cardioversion is often warranted. In addition, pharmacologic therapy with amiodarone, esmolol, verapamil, or adenosine may be used in the initial treatment of or to prevent the recurrence of supraventricular tachycardia.

The third “C” stands for CO or contractility. Contractility may be estimated from TEE or PA catheter data, if available.

The fourth “C” refers to cells (i.e., red blood cells [RBCs]). The patient’s hemoglobin concentration should be 7 to 8 g/dL, or slightly higher, before weaning from CPB. If the hemoglobin concentration is less than 6.5 g/dL when rewarming commences, the perfusionist and anesthesiologist can consider hemoconcentration or transfusion of a unit of packed RBCs (PRBCs).

The fifth “C” refers to calcium, which should be immediately available for possible administration to treat hypocalcemia and hyperkalemia. Ionized calcium levels should be measured after rewarming to help direct therapy. Although calcium is not administered routinely, if ionized calcium levels are in the low-normal range, SVR can be beneficially increased by calcium administration.

The sixth “C” stands for coagulation. After protamine is administered, ACT is measured. In patients at risk for coagulation abnormalities, prothrombin time, PTT, and platelet count should also be measured a few minutes later. If POC coagulation monitoring such as the viscoelastic tests are available, these should be measured at this time. Examples of patients at risk for coagulation abnormalities include the following: those with long CPB times; those with extreme hypothermia, elective circulatory arrest, or both, during CPB; and those with chronic renal failure. Platelet function tests may be useful in patients taking platelet inhibitors (e.g., clopidogrel or aspirin). (For further discussion of patients having emergency surgery who are taking warfarin or who have been exposed to thrombolytic drugs, antiplatelet Gp IIb / IIIa agents, or direct thrombin inhibitors see the sections on the “hematologic system” and on bleeding and coagulopathy.)

The first “V” stands for ventilation of the lungs. As CPB is discontinued, the venous outflow line is gradually occluded, and PA blood flow is gradually restored. Pulmonary ventilation and oxygenation must be reestablished, thus allowing the lungs to become the site of gas exchange again. Ideally, the lungs are initially reinflated manually, with a few sustained inflations to a peak pressure of approximately 30 cm H 2 O. If an IMA has been grafted to a coronary artery, the anesthesiologist must examine the surgical field during these inflations to ensure that the grafted artery is not overstretched, which could disrupt the distal anastomosis. Additionally, the compliance of the lungs is judged by these initial inflations, and bronchodilators can be administered if necessary. The surgeon should remove any fluid or blood from the pleural spaces and ensure that any pneumothorax is treated with a chest tube.

The second “V” refers to visualization of the heart, both directly in the surgical field (where the right-sided chambers are visible) and on the TEE, to estimate global and regional contractility. Furthermore, the degree of chamber filling (hypovolemic, euvolemic, or distended) can be estimated. In addition, one can do a final check for air within the cardiac chambers with TEE examination.

The third “V” stands for vaporizer, meaning that if volatile agents were used to ensure lack of awareness or to control blood pressure during CPB, the clinician usually reinstitutes a low dose immediately after weaning. However, because all the volatile agents decrease contractility and blood pressure, these effects can confuse the differential diagnosis of hypotension and myocardial dysfunction during weaning.

The final “V” refers to volume expanders. When all products from the pump have been exhausted and if blood transfusion is not indicated, crystalloid and albumin or hetastarch should be readily available to increase preload rapidly if necessary.

As for the letter “P” in the CVP mnemonic, Morris et al. explained that the first task “P” represents the need to be aware of predictors of adverse cardiovascular outcome. For example, preoperative low EF and prolonged CPB often predict difficulty in weaning the patient from CPB and the need for inotropic support. In addition, emergency surgery in patients with acute coronary syndrome may lead to myocardial stunning. Furthermore, inadequate surgical repair (e.g., incomplete coronary revascularization) may result in ongoing ischemia.

The second “P” stands for pressure. Calibration and rezeroing are accomplished shortly before the patient begins being weaned from CPB. Any discrepancy between the distal (usually radial) arterial pressure and the central aortic pressure should be recognized. Occasionally, the surgeon may need to insert a temporary aortic root cannula or a longer-lasting femoral arterial cannula to monitor systemic blood pressure accurately during and after the termination of CPB.

The third “P” refers to pressors, meaning vasopressors and inotropic agents that should be immediately available. A vasodilator, such as nitroglycerin, nicardipine, or nitroprusside, also should be immediately available.

The fourth “P” represents pacer because an external pacemaker should be readily available for all patients. Pacing is often needed to treat bradycardia. In patients with heart block, ideally, an AV sequential pacemaker is used to maintain the atrial kick.

The fifth “P” stands for potassium because hypokalemia may contribute to dysrhythmias, and hyperkalemia may result in conduction abnormalities. In addition, the patient’s ionized calcium level is usually checked; most clinicians have a low threshold for administering additional calcium chloride. Furthermore, magnesium (2-4 g) is usually administered before CPB is terminated. Although magnesium’s efficacy in preventing postoperative atrial or ventricular dysrhythmias has not been clearly demonstrated, hypomagnesemia is common after CPB. The risk-to-benefit ratio for administering a 2- to 4-g dose is low.

The final “P” refers to protamine. Many institutions require that protamine be uniquely packaged or kept in a nearby but separate area to ensure that the drug is not prematurely administered. (Administration during ongoing CPB is a disastrous error.) Nevertheless, it should take but a few moments to retrieve protamine when the surgeon, anesthesiologist, and perfusionist agree that it is time to reverse anticoagulation.

Termination of Cardiopulmonary Bypass

After all the preparations described previously have been made and the patient’s ventilation has been reestablished, the venous return to the pump is reduced by gradually clamping the venous line. The patient’s intravascular volume is carefully increased by continued inflow through the aortic or other arterial cannula. Ventricular distention should be avoided because it increases wall tension and myocardial oxygen consumption. The pump flow into the aorta is lowered, in effect moving into a “partial bypass” phase, in which some of the venous blood still goes into the pump and some passes through the right ventricle and lungs to be ejected into the aorta by the LV. Some clinicians reduce the pump flow to half flow rather than gradually reducing venous return to the pump. Once loading conditions are optimal and contractility appears adequate, the aortic inflow line may be clamped to separate the patient from CPB.

If CPB has been successfully terminated but cardiac performance is not optimal, preload can be increased by infusing additional blood from the pump into the aortic cannula, usually in 100-mL increments in adult patients. Monitoring the left intraventricular volume in a qualitative fashion by TEE, observing the right ventricle directly, and monitoring the filling pressures give a good estimate of the adequacy of preload. At this point, the anesthesiologist and surgeon jointly determine whether myocardial filling and performance are adequate. This determination can best be accomplished by using TEE to observe the global and regional function of both ventricles. Supplemental information can be obtained by measuring CO, if possible. Afterload can also be optimized at this point. Usually, 95 to 125 mm Hg is a desirable systolic pressure in adult patients in the immediate postbypass period, whereas increased systolic blood pressure should be avoided to prevent excessive stress on suture lines in the heart and aorta. If the patient is hemodynamically unstable and additional time is needed to administer initial or additional inotropes or vasoconstrictors, CPB can be reinstituted by unclamping the venous outflow line and directing all flow to the oxygenator again ( Fig. 54.11 ).

Fig. 54.11, Algorithm for weaning from cardiopulmonary bypass (CPB) . ACT, Activated clotting time; Hct, hematocrit; HR, heart rate; IV, intravenous; K + , potassium; LV, left ventricular; MAP, mean arterial pressure; Mg 2+ , magnesium; NO, nitric oxide; NPS, sodium nitroprusside; NTG, nitroglycerin; PGI 2 , prostacyclin; PH, pulmonary hypertension; RV, right ventricular; v¯ v¯ , mixed venous oxygen saturation; TEE, transesophageal echocardiography.

Once protamine is administered, the reinstitution of CPB becomes a more complicated process because the patient must first be reheparinized and antithrombin levels may be inadequate. Therefore, final checks of cardiac function, heart rate and rhythm, preload, afterload, and perfusion should be made jointly by the anesthesiologist and the surgeon. The venous cannula or cannulas are usually removed after the initial test dose of protamine is given. Many surgeons remove the aortic cannula only after at least half of the protamine dose has been administered. The rate and mode of protamine administration (incremental small boluses vs. continuous infusion) vary according to institutional and individual clinicians’ practices, but a large dose of protamine should never be administered as a rapid bolus.

Table 54.5 shows the characteristics and treatment modalities of specific TEE-diagnosed difficulties that may be encountered during weaning and termination from CPB.

Table 54.5
Characteristics and Treatment Modalities of Weaning Difficulties
From Licker M, Diaper J, Cartier V, et al. Clinical review: management of weaning from cardiopulmonary bypass. Ann Card Anaesth . 2012;15:206–223.
Surgical or Technical Failure Ventricular Dysfunction Vasoplegic Syndrome Left Ventricular Outflow Tract Obstruction
Diagnostic criteria TEE
Valvular regurgitation or stenosis
Patient-prosthesis mismatch
Paraprosthetic leakage
Intracardiac shunt
Occluded vascular graft
1. TEE
↓ Contractility of LV and RV
Dilated LV and RV
↓ Relaxation
2. Hemodynamics
↓ CO and ↓ MAP
1. TEE
Preserved ventricular contractility
2. Hemodynamics
↑ Or normal CO and ↓ MAP
TEE
Systolic anterior motion of anterior mitral leaflet
LV septal hypertrophy
Pressure gradient in LV outflow tract
Incidence 2%-6% 15%-40% 4%-20% 5%-10% after mitral valve surgery
Risk factors Team and operator’s experience, qualification
Low surgical volume
Extended disease, difficult anatomy
Age (>65 years), female sex
CHF, low LVEF
LV diastolic dysfunction
Previous MI, COPD
eGFR < 60 mL/min
Extensive CAD, left main CAD
Reoperation, emergency, combined procedure
Prolonged CPB
Preoperative therapy with ACEI or angiotensin II antagonist, β-blockers, heparin
High EuroScore Prolonged CPB
Low LVEF (<35%)
Myxomatous mitral valve
Hyperdynamic LV
Short distance between MV coaptation point and LV septum
Specific treatment Reoperation
Secondary repair or valve replacement
Shunt closure
Additional coronary bypass graft
1. Drugs
Adrenergic agonists (dobutamine, epinephrine, dopamine)
Phosphodiesterase inhibitors (milrinone)
Calcium sensitizer (levosimendan)
Systemic vasodilators (NTG, NPS)
Pulmonary vasodilators (NO, PGI 2 )
2. Electromechanical Support
Biventricular pacing
Intraaortic balloon pump
Extracorporeal membrane oxygenation
Ventricular assist device
Vasopressors
Phenylephrine
Norepinephrine
Terlipressin
Methylene blue
1. Medical
Volume expansion
Inotrope discontinuation
β-blockers
2. Surgical
Septal bulge resection
MV repeat repair or replacement
ACEI, Angiotensin-converting enzyme inhibitor; CAD, coronary artery disease; CHF, congestive heart failure; CO, cardiac output; COPD, chronic obstructive pulmonary disease; CPB, cardiopulmonary bypass; eGFR, estimated glomerular filtration rate; LV, left ventricle; LVEF, left ventricular ejection fraction; MAP, mean arterial pressure; MI, myocardial infarction; MV, mitral valve, NO, nitric oxide; NPS, nitroprusside sodium; NTG, nitroglycerin; PGI 2 , prostacyclin; RV, right ventricle; TEE, transesophageal echocardiography.

Chest Closure

As the cardiac surgery proceeds toward its conclusion a critical step is still to come—chest closure. Chest closure is important because of the hemodynamic consequences that can result from it.

Usually the surgical team would announce the closure of the chest to the anesthesia team, but the anesthesiologists should remain vigilant of the timing of chest closure even if the surgeons do not announce it.

In the immediate post-CPB period patients are generally hypovolemic. Closure of the chest would exacerbate the consequence of hypovolemia (i.e., hypotension). To prepare for chest closure, anesthesiologists administer crystalloid, colloid, or blood depending on the patient’s requirement. If chest closure causes severe hypotension surgeons should be requested to reopen the chest and wait until volume resuscitation has caught up (or at least the volume status is relatively easily manageable) to close the chest.

In addition to hypovolemia, chest closure is important in regard to causing ischemic changes by impinging on a venous or arterial graft that lies in a vulnerable position on or around the heart. In this situation ECG and/or hemodynamic changes will be observed which should prompt the anesthesiologist to inform the surgical team, who will reopen the chest and reposition the graft in a way that it is not affected by chest closure.

Other reasons for severe hypotension during chest closure in addition to hypovolemia and ischemia secondary to kinking of a coronary graft include impairment of RV contractility and venous return in patients with significant myocardial edema. TEE can be particularly useful in determining the cause of hypotension during chest closure because it can quickly reveal tamponade, hypovolemia, RV or LV global dysfunction, and severe new wall motion abnormalities. Reopening the chest may be necessary while treatment is instituted. Occasionally, the patient’s sternum cannot be closed because of hemodynamic instability; in such cases, only skin closure is attempted, and plans are made to return to the operating room for sternal wiring after a period of myocardial recovery in the ICU.

Transport to the Intensive Care Unit

Transportation of postcardiac surgical patients to the ICU is frequently dangerous and underestimated. Preparation for transportation of a postcardiac surgical patient should start with evaluation of the stability of the patient in the operating room. An ICU bed with a portable hemodynamic monitor should be prepared and ready for these patients. Monitoring should not be completely interrupted even for a few moments. The ideal transport monitoring system has a “brick” that can be ejected from the operating room monitor and is compatible with the transport monitor. If such equipment is not available, sequential disconnection of monitors is advised so that an online monitor is always visible and the patient is never “unmonitored.”

Staff should be educated about the importance of sequential transfer of monitors.

In the post-CPB period patients are frequently receiving infusions. A cardiac anesthesiologist should make sure that the pumps used for infusions in cardiac surgery are adequately functional. It is a good practice to unplug infusion pumps a few minutes before leaving the operating room to test the battery life for transport. Regardless of the proximity of the ICU to the operating room, disconnection of vasoactive infusions during transport could be devastating in certain critically ill patients.

As many patients are transported intubated, it is a good practice to carry a laryngoscope blade and an endotracheal tube. Even if the patient is extubated before leaving the operating room, airway management equipment and a means to ventilate the patient should still travel to the ICU with the patient. In addition, emergency medications should be brought during transport. It is recommended that the anesthesiologist carries at least one round of “code drugs” to assist in the event of cardiac arrest during transportation. A defibrillator should be on every patient’s transportation bed.

Upon reaching the ICU, it is the responsibility of the cardiac anesthesiologist to ensure that a detailed report is given to the receiving physician or nurses.

On arrival in the ICU or cardiac recovery area, a transfer of the patient and the patient’s information from one team to another, termed handoff or handover, occurs. Handoff failures have been identified as a significant source of medical errors, both between and within teams. Implementing a handoff protocol reduces information omission and reduces errors. The process is intended to be strictly sequential: monitoring should be transferred before ventilator transfer, and all phase 1 items should be completed before information transfer. Using formal, sequential handoff procedures does not increase the duration of the process. The following sequence is suggested (Wahr J, personal communication, November 17, 2012):

Phase 1: Equipment and Technology Handover

  • 1.

    Monitoring transferred to ICU equipment

  • 2.

    Ventilator function initiated

  • 3.

    Infusions and fluids checked

  • 4.

    Chest drains secured and on suction

  • 5.

    Vital signs confirmed to be stable, ventilator functioning well, infusions running appropriately

  • 6.

    Anesthesiologist, nurse, and surgeon confirm that they are ready for information transfer

Phase 2: Information Handover

  • 1.

    Anesthesiologist presents

    • a.

      Patient-specific information (age, weight, medical and surgical history, allergy status, baseline vital signs, pertinent laboratory results, diagnosis, current condition, and vital signs)

    • b.

      Anesthetic information (intraoperative course and any complications, lines present, blood transfusion or fluid totals, paralytics or opioids, antibiotics, current infusions, vital sign parameters or limits, pain relief plan, laboratory values)

  • 2.

    Surgeon presents: surgical course (diagnosis, operation performed, surgical findings, complications, blood loss, drains, antibiotic plan, deep vein thrombosis prophylaxis, medication plan, tests to be done, nutrition, key goals for the next 6 to 12 hours)

Phase 3: Questions and Discussion

In all cases, the anesthesiologist should remain with the patient until hemodynamic stability and overall stability are ensured.

The Postbypass Period: Common Problems After Cardiopulmonary Bypass

Awareness

The potential for the patient’s awareness must be assessed during and after CPB (see also Chapter 40 ). The incidence of this distressing complication is more frequent in cardiac operations than in other cases. Although patients may sweat during the rewarming period, usually because of perfusion of the thermoregulatory site in the hypothalamus with warm blood, sweating may also result from awareness if anesthetic concentrations are low during the period when the brain becomes normothermic. Awareness may be more likely if considerable time has elapsed since any sedative-hypnotic or narcotic has been administered, if small doses of anesthetic drugs were administered during CPB, or if the patient is young. Consideration should be given to continuing to administer a volatile anesthetic agent once pulmonary ventilation is reestablished and to administering additional sedative-hypnotic doses, an opioid, or both. Some clinicians begin infusing an anesthetic agent such as propofol or dexmedetomidine shortly after weaning the patient from CPB and continue it during and after transport to the ICU or cardiac recovery area.

Published studies support the hypothesis that the use of depth-of-anesthesia monitors such as the BIS can decrease the incidence of intraoperative awareness in patients at high risk for this problem (see also Chapter 40 ). However, falsely high BIS values during cardiac surgery have been attributed to interference from pump head rotation, pacemakers, and hypothermia itself. Furthermore, because processing of the raw EEG and data smoothing to generate the BIS number occur over 15 to 30 seconds, the BIS number lags slightly behind clinical events.

Another important decision to be made is whether additional neuromuscular blocking drugs are needed during and after weaning from CPB. Use of a peripheral nerve stimulator may facilitate this decision (see also Chapter 43 ). Although movement of the patient may serve as an indication of the patient’s awareness, such movement can be extremely dangerous if it results in dislodgment of the aortic or venous cannulas. Furthermore, shivering can occur because of the “afterdrop” in temperature after a period of hypothermic CPB. Because shivering can increase oxygen demand by 300% to 600%, it should be prevented by administering a neuromuscular blocking drug.

Cardiovascular Decompensation (Low Cardiac Output Syndrome)

Although improvements in myocardial protection have occurred during recent decades, it is well documented that significant declines in LV function after CABG and other cardiac operations occur in the first 8 to 24 postoperative hours. A combination of ischemia and reperfusion injury after cardiac surgery contributes to an energy deficit state in the myocardium that limits uptake of exogenous energy substrates from blood ( Box 54.8 ). Prolonged aortic cross-clamp time, incomplete revascularization, or poor myocardial preservation adds additional risk. In particular, patients with preexisting LV dysfunction experience a delay in myocardial recovery after cardiac surgery and require measures to relieve the workload of the heart. Furthermore, preexisting diastolic dysfunction is associated with an increased risk of difficulty in weaning from CPB and ongoing need for vasoactive support during the postbypass period and in the ICU.

Box 54.8
Risk Factors for Low Cardiac Output Syndrome After Cardiopulmonary Bypass
From Kaplan JA, Reich DL, Savino JS, eds. Kaplan’s Cardiac Anesthesia: The Echo Era. 6th ed. St. Louis: Saunders; 2011:1028.

  • Preoperative left ventricular dysfunction

  • Valvular heart disease requiring repair or replacement

  • Long aortic cross-clamp time and total cardiopulmonary bypass time

  • Inadequate cardiac surgical repair

  • Myocardial ischemia and reperfusion

  • Residual effects of cardioplegia solution

  • Poor myocardial preservation

  • Reperfusion injury and inflammatory changes

Criteria used to define low CO syndrome (LCOS) include a cardiac index of less than 2.4 L/min/m2, elevated lactate levels, and urine output of less than 0.5 mL/h for more than 1 hour.

Postoperative management of patients at high risk for LCOS requires a physiologic approach. Optimizing preload and reducing afterload help maximize cardiac function. Both tachycardia and bradycardia should be avoided, and postoperative arrhythmias should be treated. In addition, shivering should be prevented because it raises the heart rate by increasing oxygen demand. Postoperative deep sedation and muscle relaxation are often used to reduce myocardial workload by reducing the body’s overall metabolic demand by 25% to 30%.

Pharmacologic support is often needed to improve contractility as the patient is weaned from CPB and, eventually, recovering in the ICU ( Table 54.6 ). Catecholamines (β-adrenergic agonists) and phosphodiesterase inhibitors are the main classes of pharmacologic agents used for this purpose. Catecholamines (e.g., epinephrine, norepinephrine, dopamine, dobutamine, dopexamine, isoproterenol) are often the first line of therapy. They exert positive inotropic action by stimulating the β 1 receptor, which leads to increased intracellular cyclic adenosine monophosphate (cAMP). The predominant hemodynamic effect of a specific catecholamine depends upon the degree to which the α, β 1 , β 2 , and dopaminergic receptors are stimulated. Phosphodiesterase inhibitors (e.g., milrinone, amrinone), sometimes termed inodilators, may be used either as first-line therapy or added to β-adrenergic therapy. Phosphodiesterase inhibitors augment β-adrenergic stimulation by inhibiting the breakdown of cAMP. When these drugs are added to catecholamine infusions, the result is an additive or possibly synergistic increase in inotropy. Phosphodiesterase inhibitors also induce systemic and pulmonary vasodilation. Hence, they are particularly useful in patients with pulmonary hypertension, RV failure, or aortic or mitral valve regurgitation.

Table 54.6
Relative Potency of Commonly Used Vasoactive Drugs
From Hollenberg SM, Parrillo JE. Acute heart failure and shock. In: Crawford MH, DeMarco J, Paulus WJ, eds. Cardiology . 3rd ed. Philadelphia: Saunders; 2010:964.
Cardiac Peripheral Vasculature
Dose Heart Rate Contractility Vasoconstriction Vasodilation Dopaminergic
Norepinephrine 2-40 μg/min + ++ ++++ 0 0
Dopamine 1-4 μg/kg/min + + 0 + ++++
4-20 μg/kg/min ++ ++, +++ ++, +++ 0 ++
Epinephrine 1-20 μg/min ++++ ++++ ++++ +++ 0
Phenylephrine 20-200 μg/min 0 0 +++ 0 0
Vasopressin 0.01-0.03 units/min 0 0 ++++ 0 0
Dobutamine 2-20 μg/kg/min ++ +++, ++++ 0 ++ 0
Milrinone 0.375-0.75 μg/kg/min + +++ 0 ++ 0
Levosimendan 0.05-0.2 μg/kg/min + +++ 0 ++ 0

Although not yet available in the United States, a newer class of drugs—the calcium sensitizers—exhibit potent inodilatory properties. Levosimendan is the first such drug in this class, and has been studied extensively in other parts of the world. A randomized controlled trial of levosimendan in cardiac surgery in the U.S., (although it was found to be beneficial), did not meet its primary endpoint, and thus the drug has not received approval by the U.S. FDA. Its mechanism of action is that it increases myocyte sensitivity to calcium by stabilization of the calcium binding to troponin C, thus enhancing actin-myosin cross-bridging and increasing contractility. Therefore, inotropic performance is enhanced, whereas diastolic performance is preserved. Like phosphodiesterase inhibitors, levosimendan may augment inotropy without significantly increasing myocardial oxygen consumption.

RV failure may also be present in patients with LCOS, and it manifests as elevated PA and CVP pressures. Echocardiography can be diagnostic—findings include an enlarged and poorly contracting right ventricle, often with significant TR. The management of RV failure consists of ensuring adequate RV filling and maintaining adequate systemic pressures to prevent RV ischemia. Afterload reduction with agents effective in the pulmonary circulation is helpful. Milrinone may reduce PVR and improve CO. Nitric oxide and inhaled prostaglandins are selective for pulmonary vasodilation. Other measures to decrease PVR include hyperventilation (higher respiratory rate) to induce mild hypocapnia and aggressive treatment of hypoxemia and acidosis.

Right Heart Failure

Any situation where the right heart is not able to fulfill the requirements of circulation is labelled as right HF. With the development of new imaging modalities, it has become easy to accurately evaluate the right heart for management purposes.

Box 54.9 lists the key points of right HF which include the importance of identifying the problem in a timely manner with the help of new imaging modalities. A cardiac anesthesiologist can play a key role in this regard by using the three-dimensional TEE imaging. A number of parameters are used to evaluate right HF by the cardiac anesthesiologist using TEE: size of the right atrium and ventricle, RV systolic function, septal curvature, tricuspid regurgitation (TR), gradient across the RV outflow tract, and an estimation of the PA and RA pressures.

Box 54.9
Key Points of Right Heart Failure
From Haddad F, Elmi-Sarabi M, Fadel E, et al. Pearls and pitfalls in managing right heart failure in cardiac surgery. Curr Opin Anesthesiol . 2016;29:68–79.

  • Right ventricular (RV) function is associated with significant mortality

  • New echocardiographic modalities using strain appear promising in predicting RV failure

  • Monitoring the impact of RV function using near infrared spectroscopy and liver hemodynamics appear as useful intraoperative methods in adjusting therapeutic interventions and fluid management

Management of right HF starts by identifying the etiology of failure: ischemia, PE, outflow tract obstruction, air embolism, etc. Maintenance of sinus rhythm, reducing RV afterload, and inotropic support play an important role in supporting the right heart. Importance of maintaining adequate MAP should not be underestimated in these cases.

Inhaled vasodilators are also used in right HF. In some institutions two inhaled agents are instituted with different mechanisms of action, especially in unexpected right HF cases.

Fluid management in these patients should be very judicious as they might already be congested.

Finally, mechanical support for the failing right heart is available and has seen significant advancement in the last few years. Depending on the situation of the patient, these devices can be either temporary or permanent in nature.

Right Ventricular Dysfunction or Failure

RV dysfunction or failure may also occur after CPB, usually because of inadequate myocardial protection, inadequate revascularization with resultant RV ischemia or infarction, preexisting pulmonary hypertension, intracoronary or pulmonary air embolism, chronic mitral valve disease, or TR. Such RV failure may be evidenced by RV distention and hypokinesis on TEE, as well as by elevations in CVP and PA pressure (PAP).

Therapy for RV failure includes increasing preload and inotropic support; milrinone, dobutamine, and isoproterenol are the usual first-line drugs. Other pharmacologic drugs occasionally used to induce pulmonary vasodilation include nitroglycerin and nitroprusside. One potential problem with the use of intravenous inodilator and vasodilator agents is that their effects are not limited to the pulmonary circulation. SVR must be adequate to maintain RV perfusion pressure. Inhaled drugs such as nitric oxide, epoprostenol (Flolan), and inhaled iloprost are considered in refractory cases. Adjuncts to decrease PVR include hyperventilation (higher respiratory rate) to induce mild hypocapnia, and preventing hypoxemia and acidosis. Rarely, patients may require support with an RVAD.

Vasoplegia

Inappropriate vasodilation with a low SVR is another common cause of immediate post-CPB cardiovascular decompensation that may result in unacceptable hypotension. Predisposing factors include long-term administration of medications such as ACEIs or angiotensin receptor blockers (ARBs), prolonged duration of CPB, severe anemia with decreased viscosity, acid-base disturbances, and sepsis. Treatment with infusion of a vasoconstrictor drug such as phenylephrine, norepinephrine, vasopressin, or, rarely, methylene blue or B12 is usually successful.

Dysrhythmias

Normal sinus rhythm is ideal because it provides an atrial contribution to ventricular filling and a normal synchronized contraction of the ventricle. However, either supraventricular or ventricular arrhythmias can occur in the immediate postbypass period.

Postoperative arrhythmias are frequently seen in postcardiac surgical procedures. Table 54.7 summarizes causes and treatments of common postoperative arrhythmias. Postoperative arrhythmias can be divided into atrial and ventricular arrhythmias.

Table 54.7
Postoperative Rate and Rhythm Disturbances
Modified from Kaplan JA, Reich DL, Savino JS, eds. Kaplan’s Cardiac Anesthesia: The Echo Era . 6th ed. St. Louis: Saunders; 2011:1030.
Disturbance Usual Causes Treatments
Sinus bradycardia Preoperative or intraoperative β blockade Atrial pacing
β-Agonist
Anticholinergic
Heart block (first, second, and third degree) Ischemia
Surgical trauma
Atrioventricular sequential pacing
Catecholamines
Sinus tachycardia Agitation or pain Sedation or analgesia
Hypovolemia Volume administration
Catecholamines Change or stopping of drug
Atrial tachyarrhythmias Catecholamines Change or stopping of drug
Chamber distention
Electrolyte disorder (hypokalemia, hypomagnesemia)
Treatment of underlying cause (e.g., vasodilator, give K + /Mg 2+ )
Possible need for synchronized cardioversion or pharmacotherapy
Ventricular tachycardia or fibrillation Ischemia
Catecholamines
Cardioversion
Treatment of ischemia; possible need for pharmacotherapy
Change or stopping of drug
K + , Potassium; Mg 2+ , magnesium.

Atrial Arrhythmias

Atrial fibrillation (AF) is the most common postoperative arrhythmia (27%-40%). Patients are at highest risk for new-onset atrial fibrillation 2 to 3 days after cardiac surgery. This arrhythmia can potentially prolong the patient’s hospital stay and increase management costs by causing hemodynamic compromise or thromboembolic complications in the postoperative period.

Many potential risk factors have been studied in efforts to predict the occurrence of postoperative atrial fibrillation. With advancing age, dilatation of the atrium interrupts cell-to-cell electrical coupling between atrial muscle fibers. Other preoperative risk factors for postoperative atrial fibrillation include a history of atrial fibrillation, chronic obstructive pulmonary disease, valve surgery, and postoperative withdrawal of a β-blocker or ACEI.

Increased preoperative hemoglobin A1c, physical activity of low intensity during 1 year prior to surgery, Caucasian race, obesity, and electrolyte disturbances (hypokalemia, hypomagnesemia) have also been shown to contribute towards increased risk of atrial fibrillation.

Perioperative factors include inadequate atrial protection during surgery, pericardial inflammation, autonomic imbalance during the postoperative period, change in atrial size with fluid shifts, electrolyte (potassium and magnesium) abnormalities, and excessive production of catecholamines. Reduced risk has been associated with the postoperative administration of β-blockers, ACEIs, potassium supplementation, and nonsteroidal antiinflammatory drugs (NSAIDs).

Treatment for atrial fibrillation includes both pharmacologic agents and electrical stimulation. Many studies have shown that β blockade significantly reduces the incidence of postoperative atrial fibrillation and that withdrawal of β-blockers increases risk. Synchronized cardioversion is reserved for patients who show signs of hemodynamic instability during atrial fibrillation. In the absence of hemodynamic instability, pharmacologic agents are used with the goal of preventing a rapid ventricular response. Drugs usually employed for this purpose include calcium channel blockers, β-blockers, magnesium, and amiodarone. Expert consultation should be obtained before treatment is initiated, especially in a stable patient.

Ventricular Arrhythmias

Although ventricular arrhythmias occur after cardiac surgical procedures, sustained ventricular arrhythmias are relatively uncommon. Associated factors may include hemodynamic instability, electrolyte abnormalities, hypoxia, hypovolemia, ischemia or infarction, acute graft closure reperfusion, and the use of inotropic agents.

Ventricular arrhythmias can range from simple premature ventricular complexes (PVCs) to VT. Simple PVCs do not pose a significant risk of life-threatening ventricular arrhythmia. Conversely, complex ventricular arrhythmias, including both frequent PVCs (>30/h) and nonsustained VT, may make patients prone to sudden death, especially in the long term. Sudden death is even more likely if ventricular function is also compromised. A study of 126 patients with postoperative complex ventricular ectopy found a mortality rate of 75%. Patients with sustained ventricular arrhythmias have a poor prognosis in both the short and the long term.

Although hemodynamically unstable VT should be treated by synchronized cardioversion, patients with PVCs or short runs of nonsustained VT with hemodynamic stability do not need to be treated. Any reversible causes should be sought and corrected. Amiodarone is reserved for hemodynamically stable patients with VT or an uncertain rhythm. Ventricular fibrillation should be treated promptly with electrical defibrillation. For long-term management of ventricular arrhythmias, in addition to antiarrhythmic agents, electrophysiologic studies or placement of an ICD should be considered.

Bradyarrhythmias

Bradyarrhythmias are not uncommon in the immediate postoperative period. In most cases, a temporary epicardial pacemaker is sufficient. In a small percentage of patients, a permanent pacemaker may be necessary, especially in patients with sinus node dysfunction or AV conduction disturbances after either CABG or valve repair. Patients who need a permanent pacemaker may receive either a single-chamber or a dual-chamber pacemaker. Multiple factors dictate which particular device would most benefit an individual patient.

Hypertension

Immediately after cardiac surgery, the patient remains prone to hemodynamic instability, including hypertension. Causes of postoperative hypertension are often multifactorial and may include withdrawal from preoperative antihypertensive medications (e.g., β-blockers and centrally acting α 2 -agonists), pain, hypoxemia, hypercarbia, and hypothermia. However, arterial vasoconstriction usually plays a central role in acute postoperative hypertension. The hazards of untreated postoperative hypertension include increased myocardial work and oxygen consumption, MI, rhythm disturbances, cerebrovascular accidents, increased bleeding, and even suture line disruption. In the postoperative period, deepening sedation to control a hypertensive episode may not be the best or the only possible approach, particularly if early extubation (fast tracking) is desirable.

Several pharmacologic agents are available for use as antihypertensive agents ( Box 54.10 ). The drugs most commonly used in clinical practice are the nitrovasodilators and the dihydropyridine-type calcium channel blockers. Because of its antiischemic effects and familiarity, nitroglycerin is often the first agent used to treat hypertension in patients who have undergone coronary revascularization. However, nitroglycerin is not always effective in such patients because it primarily causes venodilation rather than arterial dilation. In addition, patients tend to develop tolerance to nitroglycerin.

Box 54.10
Vasodilators Available for the Treatment of Perioperative Hypertension
From Levy JH. Management of systemic and pulmonary hypertension. Tex Heart Inst J. 2005;32:467–471.

  • Adenosine

  • α 1 -Adrenergic antagonists

  • α 2 -Adrenergic agonists

  • Angiotensin-converting enzyme inhibitors (enalaprilat)

  • Angiotensin II antagonists

  • Atrial natriuretic peptide (nesiritide)

  • β 2 -Adrenergic agonists

  • Dihydropyridine-type calcium channel blockers

    Intravenous vasoactive therapies in widespread use to treat perioperative hypertension.

  • Dopamine agonists

  • Hydralazine

  • Nitrovasodilators

  • Phosphodiesterase enzyme inhibitors

  • Prostaglandins

Because arterial vasoconstriction plays an important role in the development of hypertension after cardiac surgery, the therapeutic agent chosen should usually be one that effectively reduces this vasoconstriction. Sodium nitroprusside, a nonspecific venous and arterial vasodilator, is a common choice. Though the risks of coronary steal with nitroprusside are theoretical, patients with renal failure eliminate sodium nitroprusside more slowly than normal, which makes them vulnerable to the toxic effects of this drug’s metabolites (cyanide and thiocyanate).

Fenoldopam is a short-acting dopamine agonist that causes arterial-specific vasodilation by stimulating D 1 -receptors. Unlike sodium nitroprusside, fenoldopam increases renal blood flow to produce diuresis and natriuresis. Nevertheless, the results of most clinical trials regarding the renal protective effects of fenoldopam are equivocal. In addition, severe hypertension may require higher doses of fenoldopam, which may be associated with undesirable increases in heart rate.

Dihydropyridine-type calcium channel blockers, such as nicardipine and clevidipine, selectively relax arterial resistance vessels without negative inotropic or dromotropic (conduction) effects and cause generalized vasodilation of the renal, cerebral, intestinal, and coronary vascular beds. Recently, due to manufacturing issues with nitroprusside, the price in the U.S. has risen to several thousands of dollars. For this reason, the author’s institution now uses nicardipine as a first-line drug to treat hypertension.

It is important to ensure that the patient’s intraarterial pressure is monitored adequately when any vasoactive agent is administered. Vasoconstriction or poor perfusion of the extremities may create a discrepancy between central aortic and peripheral arterial pressures.

While monitoring a patient’s blood pressure it is important to take note of the position of the arterial line transducer. A transducer placed lower than the mid axillary line will give artifactually high blood pressure readings. In addition, a short radial arterial catheter may be “positional” if hand positioning is suboptimal, or “dampening” of the tracing may result from poor perfusion of the distal extremities. Occasionally, during the perioperative period, the cardiac anesthesiologist or surgeon must replace a distal peripheral arterial catheter (e.g., with a femoral arterial catheter) to ensure that the effects of vasoactive therapy are monitored accurately.

Renal Insufficiency

Perioperative renal failure that necessitates dialysis occurs in approximately 2% of patients. Although the definition of renal insufficiency or failure varies among studies, three useful criteria are (1) a serum creatinine level more than 44 mmol/L (>0.5 mg/dL) higher than its preoperative value, (2) a serum creatinine level more than 50% greater than its preoperative value, and (3) a serum creatinine level more than 177 mmol/L (>2.0 mg/dL). Another definition of acute renal insufficiency involves a classification scheme that uses the acronym RIFLE ( Table 54.8 ).

Table 54.8
RIFLE Classification Scheme for Acute Renal Failure
From Kuitunen A, Vento A, Suojaranta-Ylinen R, et al. Acute renal failure after cardiac surgery: evaluation of the RIFLE classification. Ann Thorac Surg . 2006;81:542–546.
GFR Criteria Urine Output Criteria
Risk Plasma creatinine increased 1.5× or GFR decrease >25% <0.5 mL/kg/h × 6 h
Injury Plasma creatinine increased 2× or GFR decrease >50% <0.5 mL/kg/h × 12 h
Failure Plasma creatinine increased 3×, acute plasma creatinine ≥350 μmol/L, or acute rise ≥44 μmol/L <0.3 mL/kg/h × 24 h or anuria × 12 h
Loss Persistent acute renal failure = complete loss of kidney function >4 weeks
ESKD End-stage kidney disease (>3 months)
ESKD, End-stage kidney disease; GFR, glomerular filtration rate.

Acronym for risk, injury, failure, loss, and end-stage kidney disease.

Preoperative risk factors that are commonly associated with postoperative renal dysfunction after cardiac operations include preexisting renal insufficiency, type 1 diabetes mellitus, age older than 65 years, major vascular surgery, arteriopathy, genetic predisposition, and recent exposure to nephrotoxic agents (e.g., radiocontrast dyes, bile pigments, aminoglycoside antibiotics, and NSAIDs). Ejaz and colleagues showed that in addition to serum creatinine, serum uric acid is a significant predictor of AKI. In addition, several intraoperative factors may predispose a patient to renal dysfunction, including the need for emergency surgery, repeat cardiac surgery, valve surgery, and a CPB time exceeding 3 hours. Other perioperative risk factors for renal dysfunction after cardiac surgery are hypovolemia, hypotension resulting from either hypovolemia or LCOS, and embolic phenomena. Furthermore, damage to nephrons in the medullary region of the kidney results in acute tubular necrosis; hypoxia is a common cause of damage to the nephrons in this region.

Postoperative renal insufficiency in cardiac surgical patients is associated with longer ICU stay, longer overall hospital stay, and increased mortality. Thus, this disorder should be prevented whenever possible. On the basis of studies conducted in patients with radiocontrast nephropathies, investigators theorized that hydration before radiocontrast media are administered may protect the kidneys. Because the mechanism of renal injury after CPB appears similar to that triggered by the administration of radiocontrast dyes, it is thought that adequate hydration and maintaining normovolemia can help to prevent postoperative renal dysfunction in cardiac surgical patients.

Several treatment modalities have been suggested to prevent or ameliorate postoperative renal dysfunction ( Box 54.11 ). Basic supportive therapy involves ensuring adequate CO, perfusion pressure, and intravascular volume. Discontinuing any nephrotoxic drug (NSAIDs, certain antibiotics) is usual. Diuretics are not helpful and may be harmful. Unproven pharmacologic therapies include mannitol, calcium channel blockers, ACEIs, atrial natriuretic peptide, and N -acetylcysteine. Finally, if dialysis is needed, continuous dialysis may be better than intermittent dialysis.

Box 54.11
Aims of and Treatment Modalities to Reduce or Prevent the Development of Postoperative Renal Dysfunction
Modified from Sear JW. Kidney dysfunction in the postoperative period. Br J Anaesth. 2005;95:20–32.

  • 1.

    Maintain adequate oxygen delivery—by ensuring adequate cardiac output, adequate oxygen-carrying capacity, and proper hemoglobin saturation.

  • 2.

    Suppress renovascular constriction—by ensuring adequate volume preload and use of infusions of mannitol, calcium entry blockers, and angiotensin-converting enzyme inhibitors.

  • 3.

    Promote renal vasodilation—by dopaminergic agents, prostaglandins, and atrial natriuretic peptide.

  • 4.

    Maintain renal tubular flow—by loop diuretics and mannitol (which may act to prevent tubular obstruction, which can cause cellular swelling, ischemia, and death).

  • 5.

    Decrease oxygen demand—by the use of loop diuretics and mild cooling.

  • 6.

    Attenuate ischemic reperfusion injury—as a result of the release of oxygen free radicals and calcium ions.

Central Nervous System Dysfunction

The risk of overt postoperative stroke has decreased over the last few decades. However, an increased risk of neurologic complications remains in older patients, as well as in patients undergoing combined CABG and valvular heart surgery or other complex cardiac surgical procedures. Risk factors for overt neurologic complications are listed in Box 54.4 . The impact of overt stroke is profound in terms of worse adjusted hospital outcomes, longer ICU and postoperative stays, and poorer downstream survival.

A less well-defined entity, POCD or the newer terminology of PND, is far more common. Delirium is included in the definition of POCD, as are deficits of memory, concentration, and psychomotor speed. Early cognitive loss, once thought to be transient, may persist for 5 years after cardiac surgery in as many as 40% of patients. Formerly, POCD was assumed to be caused by physiologic disturbances resulting from CPB. However, more recent studies have confirmed that POCD occurs with similar frequencies after on-pump cardiac surgery, off-pump cardiac surgery, coronary stenting, and noncardiac surgery. Hence, current attention focuses on factors related to surgical stress, anesthetic agents, and patient-related predisposing factors, particularly the degree of preoperative cerebrovascular disease. In fact, surgical procedures are likely to uncover a patient’s susceptibility to cognitive loss that was already present and would eventually manifest even without surgery, usually as a result of the progression of cerebrovascular disease. Although POCD is less devastating than stroke, its potential impact on quality of life and overall healthcare resource use is still profound.

Neuroprotection Strategies

Numerous strategies have been tried to decrease the incidence and severity of neurologic injury in cardiac surgical patients. The most common nonpharmacologic approaches emphasize the reduction of macroemboli and microemboli. As described in earlier sections of this chapter, these strategies include avoiding aortic atheroma by using transesophageal or epiaortic echocardiography, optimizing the placement of the aortic cannula in the aorta, avoiding partial occlusion clamping of the aorta for proximal anastomoses by using a single cross-clamp, and, in selected patients, avoiding all cross-clamping of the aorta (the “no touch technique”). Other strategies to minimize particulate microemboli include the routine use of arterial filtration in the cardiopulmonary circuit and use of the cell saver before reinfusing blood suctioned into the cardiotomy, to remove particulate and lipid material. Strategies to minimize air microemboli include carefully removing air after any procedure involving opening a cardiac chamber, and flooding the field with carbon dioxide to minimize embolism of air entrained into the heart from the surgical field. Eliminating CPB by performing off-pump CABG surgery has been touted as a means to reduce emboli in selected patients, but this approach has not decreased POCD rates at 1 or 5 postoperative years. Many of the late-occurring strokes are probably caused by atrial fibrillation. Early pharmacologic or electrical correction and adequate anticoagulation are mandatory (see the section on arrhythmias in the postoperative period).

Other nonpharmacologic approaches include perioperative temperature control, as well as blood gas management (α-stat or pH-stat) during hypothermic CPB. These considerations are discussed in the section on CPB.

Diabetes is recognized as a risk factor for stroke and delirium after cardiac operations. Even in nondiabetic patients, hyperglycemia is extremely common during cardiac surgical procedures because of the stress response to surgery (as well as CPB), the resultant increases in circulating catecholamines and cortisol, and hypothermia-induced reductions in insulin effectiveness. Experimental evidence has revealed a relationship between hyperglycemia and worse outcome after different types of neurologic injury. However, attenuating the hyperglycemic response to cardiac surgery or CPB has proved difficult. Furthermore, concerns exist regarding inducing even a single episode of severe hypoglycemia with aggressive attempts to control glucose levels. The aim of perioperative glucose control for most patients is a level between 140 and 180 mg/dL.

The influence of intraoperative hemodynamics may influence neurologic and other outcomes. The prospective, randomized trial conducted by Gold and colleagues compared maintaining “normal” MAP (minimum of 50 mm Hg) with keeping the MAP elevated (target of 80-100 mm Hg). These investigators found that the overall incidence of combined cardiac and neurologic complications was significantly lower in the high-MAP group. In another study, investigators noted a higher frequency of hypoperfusion-type “watershed” strokes in patients who underwent cardiac surgical procedures with a MAP that was at least 10 mm Hg lower during CPB than before CPB. The current recommendation is to maintain higher targets for MAP (i.e., >50 mm Hg) in patients at high risk for neurologic injury. Because high-risk patients can be identified and a defined therapeutic window exists, pharmacologic protection would be very desirable. However, no definitively proven drug therapy currently exists for the prevention or treatment of neurologic injury after cardiac surgery.

Cerebral oximetry has been proposed to be of benefit in preventing neurologic injury in cardiac surgery, but the evidence is still lacking in this regard.

Postoperative Management of Central Nervous System Injury or Dysfunction

Postoperative stroke may be suggested by the patient’s inability to follow commands or move all extremities shortly after the surgical procedure. Neurologic consultation should be obtained, including diagnostic imaging. Diffusion-weighted magnetic resonance imaging is the most sensitive and accurate imaging technique for postoperative cardiac surgical patients. It detects more microembolic lesions than does conventional magnetic resonance imaging and is better able to find multiple watershed lesions.

Management of CNS injury or dysfunction after cardiac operations consists of general supportive measures. Hypotension should be avoided, and consideration should be given to using volume expansion, inotropic augmentation (pharmacologic or mechanical), or vasoactive medications (vasopressin or phenylephrine) to support blood pressure and brain perfusion. The brain oxygen supply-demand balance should be optimized through adequate oxygenation, sedation, and strict temperature control. Hyperthermia (fever) should be aggressively controlled. Both hyperglycemia and hypoglycemia should be avoided. Lytic therapy is of little use in cardiac surgery because it poses a risk of postoperative surgical bleeding.

POCD can manifest subtly, being evident only with psychometric testing, or, at the other extreme, it can manifest as postoperative delirium. Delirium is defined as an acute and overt change in cognition and attention, which may include alterations in consciousness and disorganized thinking. In the cardiac surgical literature, the reported incidence of delirium depends greatly on the methods used for delirium assessment, varying from 3% (chart review only) to 8% (interviews with nurses); however, with rigorous daily mental status testing and the application of a validated diagnostic algorithm, the incidence may be as high as 53%.

Risk factors include preexisting cognitive impairment, poor preoperative functional status, prior stroke or transient ischemic attack, depression, alcohol abuse, and abnormal preoperative laboratory values (glucose, sodium, potassium, and albumin). Precipitating factors for delirium in the postoperative period include intraoperative and postoperative medications, particularly sedatives and analgesics. The postoperative environment in the ICU often results in sleep deprivation and overstimulation, contributing to the onset of delirium. Complications of hospitalization and surgical procedures, including those that result in prolonged controlled ventilation and reduced mobility, also contribute to the development and severity of delirium.

Nonpharmacologic postoperative strategies to prevent delirium are listed in Table 54.9 . Medications, including those used to control pain and anxiety, are a common cause. Debate continues in the literature for a better drug to minimize the incidence of delirium between benzodiazepines and dexmedetomidine. For patients who develop agitation, the mainstay of treatment is a thorough review of medications, as well as removing other precipitating factors, such as low CO or perfusion state, metabolic disorders (e.g., hyperglycemia), fluid and electrolyte disturbances (hypoglycemia or hyperglycemia and uremia), constipation, urinary retention, and environmental noise. For patients in whom these nonpharmacologic interventions are not sufficient, an antipsychotic, usually haloperidol, is considered first-line therapy for agitation associated with delirium.

Table 54.9
Prevention of Delirium after Surgical Procedures
Modified from Rudolph JL, Marcantonio ER. Postoperative delirium: acute change with long-term implications. Anesth Analg . 2011;112:1202–1211.
Module Postoperative Interventions
Cognitive stimulation Orientation (clock, calendar, orientation board)
Avoidance of cognitively active medications
Improvement in sensory input Glasses
Hearing aids and amplifiers
Mobilization Early mobilization and rehabilitation
Avoidance of psychoactive medication Elimination of unnecessary medications
Pain management protocol
Fluid and nutrition Fluid management
Electrolyte monitoring and repletion
Adequate nutrition protocol
Avoidance of hospital complications Bowel protocol
Early removal of urinary catheters
Adequate central nervous system oxygen delivery, including supplemental oxygen and transfusion for very low hematocrit
Postoperative complication monitoring protocol

Delirium may accelerate cognitive decline in patients with Alzheimer disease or cause a type of posttraumatic stress disorder in younger patients. The long-term mental health implications of delirium have not been fully studied but may include impairment of functional recovery.

Peripheral Nerve Injuries

Peripheral nerve injuries are not uncommon in cardiac surgical patients and are often self-limited. These are the result of positioning of the upper extremities with inadequate padding of the ulnar nerve. In addition, the brachial plexus is also prone to injury from excessive stretch by the sternal retractor. Usual presenting symptoms are numbness, weakness, pain, decreased reflexes, and diminished coordination.

Phrenic nerve, recurrent laryngeal nerve, and sympathetic chain injuries have also been reported. Saphenous nerve injury during saphenous vein harvesting is also known to have been reported.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here