Cardiovascular Anesthesiology: Review Article

Anesthetic Management During Cardiopulmonary Bypass

A Systematic Review

Barry, Aaron E. MD*; Chaney, Mark A. MD*; London, Martin J. MD

Author Information
Anesthesia & Analgesia 120(4):p 749-769, April 2015. | DOI: 10.1213/ANE.0000000000000612
  • Free

Abstract

Cardiopulmonary bypass (CPB) required for cardiac surgery presents unique challenges to the cardiac anesthesiologist responsible for providing the 3 most basic facets of any anesthetic: amnesia, analgesia, and muscle relaxation. Unique pathophysiologic changes during CPB result in pharmacokinetic alterations that impact the serum and tissue concentrations of IV and volatile anesthetics. Similarly, CPB causes pharmacodynamic alterations that impact anesthetic efficacy. The clinical significance of these alterations represents a “moving target” as practice evolves and the technology of CPB circuitry advances. In addition, perfusionists choose, modify, and maintain the CPB circuitry and membrane oxygenator. Thus, their significance may not be fully appreciated by the anesthesiologist. These issues have a profound impact on the anesthetic state of the patient. The delivery and maintenance of anesthesia during CPB present unique challenges. The perfusionist may be directly responsible for the delivery of anesthetic during CPB, a situation unique to the cardiac suite. In addition, monitors of anesthetic depth—assessment of clinical signs, hemodynamic indicators, the bispectral index monitor, end-tidal anesthetic concentration, or twitch monitoring—are often absent, unreliable, or directly impacted by the unique pathophysiology associated with CPB. The magnitude of these challenges is reflected in the higher incidence of intraoperative awareness during cardiac surgery. Further complicating matters are the lack of specific clinical guidelines and varying international policies regarding medical device specifications that add further layers of complexity and introduce practice variability both within institutions and among nations. We performed a systematic survey of the literature to identify where anesthetic practice during CPB is evidence based (or not), identify gaps in the literature to guide future investigations, and explore the implications of evolving surgical practice, perfusion techniques, and national policies that impact amnesia, analgesia, and muscle relaxation during CPB.

Clinical management of patients requiring cardiopulmonary bypass (CPB) for cardiac surgery is complex, and the cardiac anesthesiologist is faced with many challenging management issues as surgical techniques evolve, such as altered cannulation sites, minimally invasive cardiac surgery, profound hypothermic circulatory arrest, and use of mechanical assistance.1 While busy confronting these challenges, the cardiac anesthesiologist must also ensure adequacy of the 3 basic facets of any anesthetic: amnesia, analgesia, and muscle relaxation.2 Objective assessment of these facets is challenging during CPB because traditional clinical variables used to assess “adequacy” of anesthesia, such as heart rate, arterial blood pressure, respiratory rate/pattern, and end-tidal carbon dioxide tension, are either not available or are directly controlled by the perfusionist. A unique aspect of this period is that critical aspects of anesthetic management, such as volatile anesthetic delivery, are transferred entirely to, or at best comanaged with, the perfusionist.

Institution of CPB induces many complex circulatory changes (pressure, flow, distribution) and profoundly alters major organ blood flow to the brain, heart, lung, kidneys, and liver.1 Unique pathophysiologic changes accompany these alterations in circulation and blood volume, such as hemodilution, hypothermia, and the systemic inflammatory response. These alterations profoundly influence the pharmacokinetics and pharmacodynamics of IV and volatile anesthetics.3,4 This systematic review focuses initially on the mechanics of CPB and resulting pathophysiologic alterations, influencing pharmacokinetics and pharmacodynamics of commonly used classes of anesthetics. We then attempt to synthesize high-quality studies and identify gaps in the literature pertaining to the proper maintenance of amnesia, analgesia, and muscle relaxation in patients undergoing CPB. The literature search strategy is presented in the Appendix.

CARDIOPULMONARY BYPASS CIRCUITRY

The CPB circuit (Fig. 1) consists of a venous reservoir, membrane oxygenator, and centrifugal pump designed to drain venous blood and return oxygenated blood to the arterial system.5 Standard CPB circuits use roller pumps for suction, venting, and delivering cardioplegia and are typically equipped with a heat exchanger, filters, real-time blood gas monitors, pressure and flow monitors, and various safety mechanisms. CPB components may be coated with heparin, albumin, or other compounds designed to minimize damage to blood products and to potentially attenuate the systemic inflammatory response.6

F1-13
Figure 1:
The cardiopulmonary bypass circuit. Reproduced with permission from Wolters Kluwer Health.5

During CPB, oxygen and volatile anesthetic are delivered via a membrane oxygenator inflow. Carbon dioxide and waste anesthetic gas are removed via an oxygenator exhaust port. It is important to note that anesthetic vaporizers are added to the CPB circuit by perfusionists after the equipment has been purchased and without any guidance from the manufacturer.7 This lack of standardization in anesthetic vaporizer integration into the CPB circuit may contribute to substantial variability in volatile anesthetic delivery within and between hospitals.

Differences in oxygenator design also influence volatile anesthetic delivery. Table 1 presents the 2 types of membrane oxygenators in common use. Microporous polypropylene (PPL) hollow-fiber membrane oxygenators function via capillary action and countercurrent gas exchange (Fig. 2).8,9 The hollow fibers are fragile, and these oxygenators are only rated by the Food and Drug Administration for a maximum of 6 to 8 hours of use.10,11 Longer durations may be associated with a decreased ability to oxygenate the patient and membrane “weeping,” which usually requires brief separating from CPB to replace the oxygenator. Consequently, PPL membranes are best suited for short-term applications. Although these membranes are the standard oxygenators used during CPB in the operating room, the risk of gas embolism also limits its safety.9 Poly-(4-methyl-1-pentene) (PMP) membrane oxygenators were developed more recently to address some of the limitations of PPL oxygenators. They are also called plasma-tight or diffusion oxygenators.12 The clinical advantages include stability for more than 6 to 8 hours, less risk of gas microembolism, and less plasma leakage across the membrane.11 Given their stability, PMP oxygenators are best suited for long-term applications, and they are used mainly during prolonged extracorporeal membrane oxygenation. Clinical ramifications of differences in CPB circuit or membrane oxygenator design are considered later.

T1-13
Table 1:
Two Types of Membrane Oxygenators in Common Use
F2-13
Figure 2:
Membrane oxygenator. Reproduced with permission from Wolters Kluwer Health.5

PHARMACOKINETIC ALTERATIONS

Table 2 lists clinical factors influencing pharmacokinetic alterations during CPB. Previous reviews by Mets4 in 2000 and Hall3 in 2002 address these in detail. Herein, we provide a brief review and update with new information accumulated over the past decade. Table 3 presents an overview of important studies investigating pharmacokinetic changes observed during CPB.

T2-13
Table 2:
Clinical Factors Influencing Pharmacokinetic Changes Observed During Cardiopulmonary Bypass
T3-13
Table 3:
Overview of Important Studies Investigating Pharmacokinetic Changes Observed During Cardiopulmonary Bypass

IV ANESTHETICS

Effects of Hemodilution

Hemodilution is a consequence of crystalloid or colloid volume priming of the CPB circuit. This decreases blood concentrations of all IV and volatile anesthetics on initiation of CPB.13–24 The choice of priming fluid may be important because anesthetics are less soluble in crystalloid and albumin than in blood.25,26

The amount of prime impacts degree of anesthetic dilution. Techniques to reduce transfusion, such as autologous priming,27–30 aim to minimize hemodilution by restricting volume of crystalloid or colloid. Similarly, minimal extracorporeal circulation (MECC) is a new technology that aims to reduce transfusion requirements and potentially attenuate the systemic inflammatory response.31,32 In short, MECC uses smaller CPB circuits (less blood-circuit interface), which require smaller priming volumes. “MECC” is a generic term, and each MECC circuit setup varies among individual perfusionists. MECC is not the standard of care in the United States because of the numerous clinical disadvantages of this technique, which include decreased safety (many safety devices are eliminated), inability to manipulate large volumes of fluid in and/or out of the patient, and sometimes lack of a volatile anesthetic vaporizer. Autologous priming and MECC (typical prime approximately 400 mL, in contrast to “standard” prime of 750–1000 mL) both decrease the volume of prime solution and may attenuate the effect of hemodilution on anesthetic blood concentrations. However, these techniques have only recently been introduced. Thus, their impact on anesthetic blood concentrations during CPB has not been adequately studied. Essentially, all the literature forming the basis of our understanding of the effects of hemodilution was published during the era of standard priming volumes (750–1000 mL). However, one study demonstrated that smaller priming volumes (300–360 mL vs 750–1000 mL) resulted in less hemodilution of fentanyl during pediatric cardiac surgery.33

Altered Plasma Protein Binding

Priming volume at onset of CPB decreases blood concentrations of albumin,17,18,33α1-acid glycoprotein,13 and red blood cells,3,14,25 all of which bind anesthetics. This is of particular relevance for IV drugs that are highly protein bound, with high hepatic extraction ratios or low therapeutic indices.34 Consequently, propofol, midazolam, and opioids exhibit increased free fractions of drug during CPB.17,18,20,22,33 The free unbound component of drug is what diffuses across the blood-brain barrier, producing anesthetic effect.35–37 The increased free fraction offsets the decrease in overall drug concentration from hemodilution (Fig. 3). Several studies have revealed that unbound drug concentrations are unaffected by initiation of CPB and in many cases may exceed pre-CPB levels, despite significant decreases in total drug concentration.18,20,22 Decreased hepatic and renal clearance may also contribute to higher levels of unbound drug.38,39 Thus, anesthetic depth may be preserved or even enhanced, despite decreases in total anesthetic blood concentration. Finally, given the substantial proportion of critically ill patients undergoing cardiac surgery, it is also important to consider the potential pharmacologic impact of hypoalbuminemia on concentrations of active, unbound drug. Higher levels of unbound propofol have been observed in critically ill patients with hypoalbuminemia,40 possibly resulting in decreased propofol requirements (Fig. 4).

F3-13
Figure 3:
Interplay of temperature, hemodilution, plasma protein concentration, heparin, and protamine on bound and unbound propofol concentrations and parameters indicating anesthetic depth. Plasma total and unbound propofol concentration, unbound fraction of propofol-packed cell volume (PCV), serum albumin concentration and latency of the Nb wave from the auditory-evoked potentials (mean [95% confidence interval]). n = 15 patients, unless otherwise indicated (1 n = 14, 2 n = 12). NA = not available. *P < 0.05 versus value awake; †P < 0.05 versus value before heparin; ‡P < 0.05 versus value before cardiopulmonary bypass; §P < 0.05 versus hypothermic cardiopulmonary bypass (CPB). Reproduced with permission from Oxford University Press.23
F4-13
Figure 4:
Unbound propofol concentrations are higher when patients are hypoalbuminemic. The percentage of free propofol in serum from control subjects and from critically ill patients; control group 1.07 (0.49–1.47%); critically ill group 1.31 (1.06–2.25%). Reproduced with permission from John Wiley and Sons.40

Heparin, particularly in the large amounts required for CPB, also affects free drug blood concentrations by initiating the release of lipoprotein lipase and hepatic lipase, each of which hydrolyzes plasma triglycerides into nonesterified fatty acids.4,41 These bind competitively to plasma proteins, displacing protein-bound drugs and increasing free fraction of drug. This effect of heparin has been well described for many drugs,42–46 and amounts as low as 50 International Units have been shown to increase the free fraction of propranolol.45 This principle also likely applies to protein-bound anesthetics, with increasing free fractions potentially deepening anesthetic level. However, many of these studies were performed decades ago, and the clinical impact of heparin administration on modern anesthetics is unclear. Of note, protamine reverses heparin’s impact on unbound drug blood concentrations.23

Volume of Distribution

Drugs with a high volume of distribution such as propofol and fentanyl distribute widely throughout blood and tissues. A high volume of distribution likely attenuates the clinical impact of hemodilution on anesthetic blood concentrations because drug distributed into tissues will rapidly re-equilibrate with plasma concentrations after initiation of CPB. Re-equilibration occurs over the course of a few minutes and has been demonstrated for many anesthetic17,20 and nonanesthetic47,48 drugs.

Altered Drug Clearance

Drug clearance is profoundly affected by hypothermia. Animal studies reveal that enzymatic efficiency is impacted by changes in temperature.49,50 In humans, hypothermia-induced decreased Hoffman elimination occurs with cisatracurium51,52 and remifentanil.22 Degree of hypothermia also impacts rate of drug clearance.52,53 A decrease in body temperature of as little as 2°C may double the duration of neuromuscular blockade.54

Over the years, substantial change in the clinical management of hypothermia during CPB has occurred, with a shift toward “mild” hypothermia (“drifting” to 32–34°C range) and, to a lesser degree, normothermic CPB.55 Much of the literature forming the basis of our understanding of the pharmacokinetic impact of CPB was published before this change in practice (e.g., at colder temperature). In addition, the terms “mild,” “moderate,” “deep,” and “profound” hypothermia are often vaguely defined and vary across studies. Thus, the effects of hypothermia on drug clearance in contemporary clinical practice may not be as significant as initially reported at colder temperatures.

Drug clearance is also affected via altered organ perfusion/microcirculatory flow during CPB. Blood flow to the liver and kidney is decreased by hypothermic CPB.3,4 This is particularly relevant for drugs with high liver extraction ratios, such as propofol.13 However, these conclusions are based on data extrapolated from animal studies56,57 and indirect measurements of hepatic function in humans.58–60 Several studies have challenged these assumptions and suggest that normothermic and mild to moderate hypothermic CPB may have only minimal effects on hepatic blood flow and function.61–64 Thus, the clinical impact of contemporary temperature management practices on hepatic and renal perfusion/function may be less than previously thought.

Drug clearance after CPB is also affected, as prolonged elimination half-lives and elevated blood drug concentrations of propofol13,65 and midazolam66 have been reported. While the mechanism is unclear, reduced elimination resulting from hypothermia and/or altered organ perfusion/microcirculatory flow are commonly cited as potential causes.59

Acid-Base Disturbances

Initiation of nonpulsatile hypothermic CPB alters microcirculatory blood flow, potentially creating physiologic areas of hypoperfusion and acidosis.67 Basic drugs may become trapped in acidic cellular microenvironments and recirculate once perfusion improves and acidosis resolves.4 Thus, during hypothermic CPB, there is the potential for “trapping” of basic drugs and their “release” after separation from CPB. This hypothesis is based on an old study of deltoid muscle gas tensions demonstrating decreased perfusion of muscle during hypothermic CPB,67 and more current research on alterations in microvascular deltoid muscle blood flow during cardiac surgery supports these findings.68 However, neither of these investigations actually confirmed that basic drugs were trapped in these cellular microenvironments because they did not measure muscle drug concentrations.

Lung Isolation

Pulmonary blood inflow almost completely ceases during CPB yet continues via bronchial artery blood flow; thus the lungs are almost isolated from the systemic circulation. Opioids and neuromuscular blocking drugs are highly sequestered in the lungs during this period and are not metabolized.23,69–71 Consequently, resumption of ventilation and reinitiation of pulmonary blood flow allow these drugs to recirculate, and increased serum concentrations of fentanyl20 and sufentanil19 have been demonstrated during separation from CPB (Fig. 5).

F5-13
Figure 5:
Increased sufentanil concentration when coming off bypass as a consequence of pulmonary arterial inflow and ventilation. Plasma sufentanil concentrations in all 4 groups. All measurements represent the mean ± SEM for 10 patients except for the last sample, 4 hours (240 minutes) after cardiopulmonary bypass. Reproduced with permission from Wolters Kluwer Health.71

Drug Sequestration by the Cardiopulmonary Bypass Circuit

Opioids,72 propofol,73–76 and volatile anesthetics8 bind tightly to the plastic components of the CPB circuit and membrane oxygenator. As much as 75% of administered propofol is absent from plasma after 120 minutes of a simulated in vitro CPB run.75 However, CPB circuits are now routinely “coated” with a variety of substances such as heparin,73 which decreases platelet adhesion, and phosphorylcholine,77 which may more closely mimic the vascular wall, in an attempt to decrease damage to blood elements and attenuate the systemic inflammatory response. In vitro analyses on the impact of these coatings on propofol have not demonstrated any effect on pharmacokinetics.23,77 There is a growing list of new “biocompatible circuit coatings”78–81 with unknown pharmacologic implications. Thus, additional advances in CPB technology may affect pharmacokinetics. Also, the amount of drug sequestration would be expected to decrease with MECC because of smaller circuit surface area and less blood-circuit interface; however, this has yet to be studied.

Systemic Inflammatory Response

CPB induces a profound systemic inflammatory response, leading to dysregulation of cytokines, interleukins, and the cytochrome P-450 enzymatic system responsible for liver drug clearance. In addition, downregulation of enzymatic systems located in the brain may reduce drug clearance and prolong anesthetic action centrally.3 MECC, corticosteroids,82 complement inhibitors,83 and other techniques used to reduce inflammation may impact anesthetic pharmacology by such attenuation, yet this has not been formally studied. The systemic inflammatory response induced by CPB is a broad topic beyond the scope of this review, and the reader is referred elsewhere.84–86 In general, the clinical impact of individual inflammatory mediators is poorly understood.3 By extension, the sum effect of the inflammatory response on pharmacokinetics is even less well defined. Interestingly, a recently published small (n = 14 animals) porcine model study hints at the potential ability of anesthetic technique to alter the systemic inflammatory response.87 Pigs administered isoflurane, in contrast to propofol, during CPB exhibited a greater increase in fluid extravasation from the intravascular to the interstitial space, resulting in dilution of interstitial fluid and a decrease in interstitial colloid osmotic pressure, implying that perhaps propofol is better than volatile anesthetics at systemic inflammatory response attenuation during CPB.

VOLATILE ANESTHETICS

The use of volatile anesthetics during CPB was first described in 1974.88 In addition to the factors discussed above, the pharmacokinetics of volatile drugs are impacted by 3 additional factors: altered blood/gas partition coefficient,24,26,89–96 altered tissue solubility,90,97,98 and oxygenator design.8,9,12,89,97,99 An important consideration when evaluating clinical studies in this arena is how the volatile anesthetic was measured. One method measures plasma concentration, which requires storage at controlled temperature and transport to a laboratory. Rapid results are not available, preventing applicability for intraoperative management. Another method is to sample the membrane exhaust port (end-tidal anesthetic concentration [ETAC]), which may offer real-time data to help guide intraoperative management.

Altered Blood/Gas Partition Coefficient

Hemodilution and hypothermia are competing physiologic factors influencing volatile anesthetics by virtue of their opposite effects on the blood/gas coefficient (hemodilution decreases, hypothermia increases). During hypothermic CPB, they act to counterbalance each other.90 During rewarming, however, the blood/gas partition coefficient decreases.91 Regarding desflurane and isoflurane, substantial increases in temperature during rewarming (30.4–37.5°C), with small increases in hematocrit (21%–22%), resulted in the lowest blood/gas partition coefficient measured (approximately 75% of preinduction levels for all 3 drugs).91 This results in more rapid washin or washout of volatile anesthetic during separation from CPB and implies that anesthetic titration (changing levels) during hypothermia is slowed.100 However, slower titration of volatile anesthetic can be partially overcome by increasing the fresh gas flow in the CPB circuit.17,99 Practice variations regarding hemodilution and temperature management strategies will impact blood/gas partition coefficients of volatile anesthetics.

Altered Tissue Solubility

Hypothermia increases solubility of volatile anesthetics in tissues, such as the brain, resulting in increased anesthetic depth.4 However, with alterations in the blood/gas partition coefficient noted earlier, titration (changing levels) of volatile anesthetics occurs more slowly during hypothermic conditions,89,90,97,101 and it may take more time to reach a deeper anesthetic state.

Oxygenator Design

Oxygenator design profoundly affects anesthetic uptake. Oxygenators either absorb volatile anesthetic8,99 or prevent passage of the agent altogether (Fig. 6).9,12 In vitro comparison of PPL and PMP membranes confirms that while volatile anesthetics cross the PPL membrane, virtually no volatile anesthetic diffuses across the PMP membrane.9 This finding has been confirmed using gas chromatographic measurement of blood isoflurane concentrations in vivo.12 While PMP oxygenators are widely used for extracorporeal membrane oxygenation given their superior stability to fragile PPL oxygenators, they may also be used during CPB in the operating room if total IV anesthesia is the planned technique, which is common throughout Europe. Despite such issues, manufacturers of membrane oxygenators (PPL or PMP) are not required to test the impact of their design on anesthetics; they are only required to ensure biocompatibility with membrane components.10 Consequently, the impact of membrane oxygenators on anesthetics may not be apparent until after the device comes to market.11

F6-13
Figure 6:
Isoflurane does not cross poly-(4-methyl-1-pentene) (PMP) membrane oxygenators. Isoflurane blood concentrations (Cisoflurane [μm]) for the uptake and elimination sequence in the 4 oxygenator groups. Each line represents a single patient. (A) Capiox®RX25; (B) Hilite®7000; (C) QuadroxD; (D) Hilite®7000LT.12 PPL = polypropylene. Reproduced with permission from Wolters Kluwer Health.12

PHARMACODYNAMIC ALTERATIONS

We are still far from a complete understanding of how anesthetics initiate and maintain the anesthetic state. This fact limits our knowledge of pharmacodynamic alterations induced by CPB. Furthermore, the relative impact of hypothermia alone or CPB on pharmacodynamic alterations cannot always be separated. Altered pharmacokinetics due to hypothermia or CPB and lack of an objective assessment of adequacy of anesthesia during this time also confound evaluation of anesthetic depth. Despite these issues, several studies indicate that hypothermia and impaired cerebral function resulting from pathophysiology associated with CPB play major roles in the pharmacodynamics of IV and volatile anesthetics (Table 4).

T4-13
Table 4:
Clinical Factors Influencing Pharmacodynamic Changes Observed During Cardiopulmonary Bypass

HYPOTHERMIA

Hypothermia clearly impacts anesthetic requirements. Studies on the effect of temperature reveal that less propofol is required to maintain a constant bispectral index (BIS) level during normothermic CPB.102 Patients exposed to hypothermic CPB require even less propofol,23,103 suggesting that hypothermia increases its potency.14,15 Studies on volatile anesthetics assessing anesthetic depth with electroencephalography or ETAC report decreased anesthetic requirement during hypothermia15,104,105 and increased anesthetic requirement during rewarming.106,107 This has been consistently demonstrated in adult patients for sevoflurane106 and isoflurane,107,108 in pediatric patients,108,109 and in animals.110 One investigation in children measured isoflurane minimum alveolar concentration requirements and isoflurane lipid solubility and suggested that anesthetic solubility alone is not enough to explain the decreased minimum alveolar concentration requirement observed during cooling. These investigators postulate that their results imply a potential amnestic effect of hypothermia.108

Hypothermia induces both pharmacokinetic and pharmacodynamic alterations. Some investigators have attempted to control these pharmacokinetic changes to delineate the potential impact of hypothermia alone on pharmacodynamics, by assessing isoflurane lipid solubility, not blood concentration, yet this is challenging.108 In reality, it remains quite difficult to distinguish clinically whether changes in anesthetic depth are due to altered pharmacokinetics or pharmacodynamics (or both).

Hypothermia also confounds studies that rely on BIS monitoring to assess anesthetic depth because BIS independently decreases with hypothermia103,104,111–114 and increases during rewarming.14,17,107,114 It is unclear whether these electroencephalographic changes represent the anesthetic state or are an artifact of changing temperature. In addition, the response of BIS to changes in temperature is highly variable among patients.109,115–117 This information, along with the demonstration of decreased minimum alveolar concentration requirement during hypothermia,108,110 would propose that electroencephalographic changes in response to hypothermia may indeed reflect changes in anesthetic depth, potentially lending validity to studies reliant on electroencephalographic monitoring.

Hypothermia also impacts analgesia and the density of neuromuscular blockade. Analgesia may be affected via decreased morphine binding at opioid receptors under hypothermic conditions.118 Neuromuscular blockade density is enhanced during hypothermia; muscle strength is impaired by as much as 10% with each decreasing degree Celsius in the absence of muscle relaxant.54 Whether the result of pharmacokinetic or pharmacodynamic alterations (or both), studies clearly demonstrate that decreased doses of vecuronium119,120 and rocuronium121 are required during hypothermic CPB.

IMPAIRED CEREBRAL FUNCTION

Exposure to CPB alone (with or without hypothermia) has been implicated as a reason for decreased minimum alveolar concentration requirement. While the mechanism is unclear, impaired cerebral function as a consequence of CPB has been the focus of many animal studies.110,122–126 However, such observational studies have lacked control groups and report inconsistent results. Several human studies during hypothermic CPB implicate CPB as an independent factor responsible for decreased anesthetic requirements.103,107,127 Specifically, initiation of CPB decreases the magnitude of propofol requirements, and the effect of hypothermic CPB is significantly more than that of normothermic CPB.103 Isoflurane requirements are also reduced during hypothermic CPB107 and isoflurane requirements are reduced after, compared with before, hypothermic CPB.127

One randomized, controlled trial assessed minimum alveolar concentration of rats exposed to normothermic CPB.126 Ten rats were cannulated and exposed to CPB and compared with a control group that underwent sham cannulation without CPB. Although isoflurane requirements remained unchanged in the sham CPB group, minimum alveolar concentration after 20 minutes in the CPB group was 10% lower than pre-CPB measurements and remained below baseline at the last measurement 2 hours after CPB. This is consistent with a study in goats demonstrating a 20% decrease in minimum alveolar concentration requirements after CPB exposure.110

Other investigations have sought to compare anesthetic requirements in patients during CPB and off-pump coronary artery bypass patients to determine the potential hypnotic impact of CPB. One study showed no difference in propofol requirements between CPB and off-pump coronary artery bypass patients.128 However, the off-pump coronary artery bypass procedures took significantly longer, and equivalent propofol consumption was only noted after correcting for time. More recently, other investigators have compared CPB with off-pump coronary artery bypass procedures and found that less propofol is required during CPB to maintain a constant BIS.129 However, these investigators did not measure serum propofol concentrations and were unable to account for the effects of altered pharmacokinetics due to CPB.

Although it appears that anesthetic requirements decrease during CPB, the mechanism of such an effect remains unclear. Numerous models of CPB-induced neurologic injury, such as microembolic load, cerebral edema, brain inflammation, decreased brain cytochrome P-450 activity, or increased intracranial pressure, have all been invoked to explain why anesthetic requirements may decrease.3,130–132 New avenues of study have identified surgical trauma as a potential source of exaggerated and persistent neuroinflammation, perhaps leading to postoperative cognitive dysfunction in elderly patients.133,134 While the etiology of postoperative cognitive dysfunction is certainly multifactorial, the systemic inflammatory response observed during cardiac surgery has also been used to explain the frequent incidence of postoperative cognitive dysfunction in this surgical population.135

HOW DO THESE ALTERATIONS AFFECT ANESTHETIC MANAGEMENT?

To the novice, initiation of CPB is the start of a mandatory “downtime” for anesthesiologists, when workload appears to diminish. However, the experienced cardiac anesthesiologist appreciates that initiation of CPB begins a unique period in which management of the patient’s physiologic and anesthetic state is shared with the perfusionist. Given the pharmacokinetic and pharmacodynamic alterations discussed earlier, physiologic controversies related to appropriate mean arterial blood pressure, flow rates, degree of anticoagulation, and absence of traditional clinical variables used to assess adequacy of anesthesia, such as changes in heart rate, pulsatile blood pressure, and the de facto delegation of several key anesthetic processes such as volatile anesthetic delivery to the perfusionist, this should be appreciated as a period of maintained vigilance for the anesthesiologist. Unfortunately, ensuring that the 3 basic tenets of anesthesia are met (amnesia, analgesia, muscle relaxation) during CPB remains challenging.

ENSURING AMNESIA DURING CARDIOPULMONARY BYPASS

Ensuring amnesia requires an accurate assessment of anesthetic depth, which is quite complicated with or without CPB. The use of BIS (discussed later) remains controversial. Monitoring anesthetic depth based on autonomic signs is challenging during CPB because these signs are largely absent. However, hypertension during normal CPB flows may indicate that anesthetic depth or the degree of neuromuscular blockade is inadequate. Traditional methods of measuring ETAC are also impractical because of the minimal blood flow through the lungs during CPB. Several studies suggest measuring the oxygenator membrane exhaust gas for halogenated anesthetic as a surrogate for ETAC.12,90,97,106,107,136 However, this is not a standard practice. Consequently, routine monitoring of patients during CPB cannot ensure that adequate anesthesia to prevent awareness is being delivered or to exclude equipment malfunction such as cracked tubing or damaged vaporizers. With the use of total IV anesthesia during CPB, there is no real-time monitor of IV anesthetic serum concentration. Empiric dosing regimens based on target-controlled infusions are distorted by pharmacokinetic and pharmacodynamic alterations during CPB.18,21,22,53,119,137,138 In addition, cardiac patients may not tolerate traditional amnestic doses without adverse hemodynamic consequences. Finally, practitioners may be hesitant to use appropriate doses of anesthetics while separating from CPB because of perceived depressant effects on the myocardium.139,140

Thus, considering all the above, it may not be surprising that cardiac surgery patients exhibit an increased incidence of recall when compared with other surgical cohorts.141–148 Initial reports of awareness using a high-dose opioid technique during the early days of cardiac surgery were common, and the incidence was reported to be as high as 23%.149 High-dose opioid techniques blunt hemodynamic responses traditionally used to assess anesthetic depth without ensuring amnesia.150 A significant decrease in the incidence of awareness in cardiac surgery occurred once high-dose opioid techniques were supplemented with benzodiazepines or volatile anesthetics.151 More contemporary methods using balanced anesthetic techniques based on the combination of low- to moderate-dose opioids, benzodiazepines, volatile anesthetics, or propofol infusions have decreased the incidence of awareness during cardiac surgery to 0.2% to 2.3%.141,142,144–148 This incidence remains significantly higher than the reported rate in the general surgical population.144,152

However, the 5th National Audit Project on accidental awareness during general anesthesia in the United Kingdom and Ireland published simultaneously in Anaesthesia and the British Journal of Anaesthesia may change our thinking regarding awareness. The incidence of certain/probable and possible accidental awareness cases was much lower than previously reported (approximately 1:19,000). Although cardiac cases remained “high risk” (approximately 1:10,000), most of the reports involved either brief interruption of drug delivery caused by human error or technical problems or the use of intentionally low doses of anesthetic drugs in high-risk patients, such as those with cardiovascular instability. The full report can be found at http://www.nationalauditprojects.org.uk/NAP5_home.

ANESTHETIC DELIVERY

In North America, the perfusionist typically delivers volatile anesthetic during CPB. The use of volatile anesthetic during CPB was initiated approximately 20 years ago in an attempt to decrease administration of long-acting opioids and sedatives to allow patients to emerge more rapidly from anesthesia and persists, in part, due to the presumed clinical benefits of volatile anesthetic preconditioning.153,154 An additional factor perhaps favoring volatile anesthetics is the anecdotal reporting of a propofol “biofilm” or “foam” that may develop on the oxygenator membrane after long CPB runs or during low-flow CPB, prompting fears of oxygenator failure and leading to hesitation or even refusal by many perfusionists to deliver propofol directly through the CPB circuit (Robert Groom, MS, CCP, Maine Medical Center, personal communication, December 16, 2013; Joerge Ender, MD, Leipzig Heart Center, University of Leipzig, personal communication, December 19, 2013). However, there is the potential for biofilm development independent of whether propofol is administered IV or directly into the oxygenator. Despite these concerns regarding propofol, we were unable to find any case report(s) or original investigations regarding this potential problem. In contrast, 1 in vitro study supports the use of propofol during CPB, concluding that there is no effect of propofol on gas exchange properties of membrane oxygenators, even at high doses (Fig. 7).77 This study makes no mention of a propofol biofilm or foam accumulating on the membrane oxygenator. Of note, older CPB circuits may not promote adequate mixing. Thus, propofol delivered directly into the CPB circuit may float to the top of the reservoir.155 This can be avoided by delivering propofol IV during CPB.

F7-13
Figure 7:
Gas exchange properties of polypropylene membrane oxygenators after exposure to propofol. Partial pressure of carbon dioxide after 180 minutes of circulation (4.0 L/min) and 400 mg of propofol exposure. The changes were not statistically significant.77 PO2 = partial pressure of oxygen. Reproduced with permission from SAGE Publications Ltd.77

In contrast to practice in North America, Europeans favor total IV anesthetic techniques during CPB. This preference has been driven by European Council Directive 93/42/EEC, which prohibits the routine use of volatile anesthetics while on CPB.156,157 This directive bans the supplementary incorporation of an anesthetic vaporizer into the extracorporeal circuit because this would add complementary safety and performance-related device features. France and Germany, for instance, use this directive to mandate that any modification performed on CPB circuits must pass a technical approval process to ensure device safety (Joerge Ender, MD, Leipzig Heart Center, University of Leipzig, personal communication, December 17, 2013). This becomes problematic because manufacturers of CPB circuits do not build them with vaporizers, requiring perfusionists to modify them.11 Thus, anesthesiologists in many European countries routinely administer propofol during CPB to avoid the device approval process (Joerge Ender, MD, Leipzig Heart Center, University of Leipzig, personal communication, December 17, 2013). Notably, the application of this directive to CPB circuits is variable across nations. For instance, the United Kingdom does not enforce it (Joerge Ender, MD, Leipzig Heart Center, University of Leipzig, personal communication, December 17, 2013).7 The combination of this directive and the literature regarding the presumed clinical benefits of volatile anesthetic preconditioning153,154 has prompted European practitioners to alternative approaches. For instance, one protocol uses MECC circuits to minimize drug sequestration and PMP membranes (prevent passage of volatile anesthetic) to maximize recirculation of tissue stores of sevoflurane accumulated before CPB in an attempt to achieve the potential benefits of volatile anesthetic preconditioning during CPB.156

In addition to variability in international practice, several hazards associated with the delivery of volatile anesthetic during CPB must be considered. Accumulation of toxic sevoflurane metabolites during CPB has been reported.158 Case reports also describe severe damage to the plastic components of the circuit by spillage of liquid volatile anesthetic (Fig. 8; video supplied on request).159–162Figure 9 demonstrates insertion of a vaporizer on a contemporary CPB machine well away from the plastic CPB components in an effort to reduce this potential hazard. However, there are no specific instructions from device manufacturers ensuring that vaporizers are placed in a safe position. Finally, scavenging systems should be connected to the membrane oxygenator exhaust to reduce occupational exposure to volatile anesthetic (Fig. 10).163,164 Notably, the Occupational Safety and Health Administration does not offer any recommendations for scavenging from the CPB apparatus.165

F8-13
Figure 8:
Damage to plastic components by liquid volatile anesthetic.
F9-13
Figure 9:
Recommended anesthetic vaporizer placement well away from plastic cardiopulmonary bypass components.
F10-13
Figure 10:
Membrane oxygenator gas exhaust port (indicated by arrow). This can be a source of occupational exposure to waste anesthetic gas (should be scavenged) and can also be used to monitor anesthetic gas concentration.

Guidelines from the medical community regarding occupational safety are also lacking. Several studies and organized initiatives regarding patient safety in the cardiac operating room and systematic reviews166–169 do not address occupational health and safety for operating room personnel.170 Similarly, these publications do not address safety with regard to occupational exposure to volatile drugs during CPB for health care providers. A recent anesthesiologist-led review provides commentary on equipment design and operating room layout yet does not provide design specifications for CPB circuits regarding anesthetic vaporizer placement or scavenging systems for waste anesthetic gas.171

The American Society of Extracorporeal Technology’s recently revised internal standards and guidelines for perfusion practice are the only guidelines available that recommend scavenging waste anesthetic gas and routine maintenance of anesthetic vaporizers.6 Notably absent are details regarding how vaporizers should be integrated into the circuit, scavenging system specifications, recommendations for titration of volatile or IV anesthetics, how and when anesthetic-related issues should be communicated to the anesthesiologist, monitoring adequacy of anesthetic delivery, or assessment of anesthetic depth.

THE BISPECTRAL INDEX

The BIS monitor (Aspect Medical Systems, Natick, MA) is the only U.S. Food and Drug Administration–approved monitor to assess the anesthetic state of the central nervous system. However, its use during cardiac surgery172 and in the general surgical population148 in preventing awareness is unreliable. Patients may report awareness, despite “appropriate” BIS levels (<60).143,173,174 However, in the absence of effect-site concentration monitors or a surrogate such as ETAC, the BIS monitor may have applicability in potentially reducing the incidence of awareness.141,173,175,176Table 5 outlines important studies evaluating the potential applicability of BIS in preventing awareness.

T5-13
Table 5:
Important Studies Evaluating Applicability of Bispectral Index (BIS) in Preventing Awareness

Additional potential clinical applications of BIS monitoring outside the CPB setting have recently been reviewed.149,172 These include more appropriate anesthetic dosing, potentially improving recovery times and facilitating fast-track techniques. Preventing anesthetic overdose and reducing hemodynamic instability from unnecessarily high anesthetic concentrations may also potentially decrease morbidity and mortality. In addition, the BIS monitor may have applicability as a monitor of cerebral ischemia (prevention of catastrophic neurologic events). However, BIS-guided anesthetic dosing has not been shown to decrease extubation/recovery time,177–180 improve morbidity or mortality,149,172,181–183 nor is it an effective monitor of cerebral ischemia.184–186 It may help alleviate postoperative delirium in older patients, by preventing too “deep” an anesthetic state (BIS values <20; noncardiac surgery).187–189

The American Society of Anesthesiologists Practice Advisory on Awareness and Brain Monitoring currently recommends that the decision to use a brain monitor should be made on a case-by-case basis and should not be considered the standard of care.190 In the United Kingdom, the National Institute for Health and Care Excellence recommends that BIS may be useful in guiding anesthetic dosing in patients at high risk for adverse consequences of anesthetics and that BIS is useful for total IV anesthesia.191 However, the National Institute for Health and Care Excellence also states that electroencephalographic-based monitors are not associated with a statistically significant reduction in intraoperative awareness in patients classified at high risk and that available evidence on the impact of these technologies in reducing awareness is limited.192 The American Society of Extracorporeal Technology’s recently revised internal standards and guidelines for perfusion practice recommend the routine use of cerebral oximetry yet do not address use of processed electroencephalographic monitors (BIS) or the assessment of anesthetic depth.6

END-TIDAL ANESTHETIC CONCENTRATION

When volatile anesthetics are used, ETAC may be clinically useful during CPB. This is not routine practice despite being tested and advocated by many investigators, demonstrating that monitoring of ETAC at the membrane exhaust port accurately reflects serum anesthetic concentrations.12,90,97,106,107,136 Others caution against this practice, reporting that exhaust gas concentrations do not correlate with serum concentrations.17 This may be due to room air admixture as a result of new safety mechanisms designed to prevent negative pressure across the oxygenator membrane.17 This conclusion was challenged by an accompanying editorial asserting that the protocol for measuring serum anesthetic concentration was flawed.193 While the sum of the literature advocates monitoring of ETAC during CPB, it is worth noting that evolving CPB circuit technology has the potential to impact the reliability of this practice.

ENSURING ANALGESIA DURING CARDIOPULMONARY BYPASS

While the focus of anesthetic studies during CPB has primarily been directed at amnesia, very little has been published regarding analgesia during CPB. Unlike amnesia, there is no potential monitor of “analgesic depth.”2 Lack of real-time monitoring of plasma drug concentrations forces anesthesiologists to rely on clinical signs of pain, such as tachycardia, hypertension, and tachypnea. These clinical signs are usually unreliable or absent before, during, and after CPB (pacemakers, vasoactive drugs, paralytic drugs). Thus, opioid dosing during CPB is empirically driven. In the past, high-dose opioid techniques likely provided some reliable degree of early postoperative analgesia. However, fast-track early extubation protocols using low-dose opioid supplementation are now the norm in clinical practice. Their implications for postoperative analgesia are complex and may vary with the type of opioid used, as well as adjuvant sedation used (e.g., potential opioid-sparing effects of dexmedetomidine) and other potential nonopioid drugs (e.g., nonsteroidal anti-inflammatory drugs).194,195

ENSURING MUSCLE RELAXATION DURING CARDIOPULMONARY BYPASS

Neuromuscular blockade during CPB may be desirable, yet there is no definitive evidence that it is absolutely required; therefore, there is wide variability in the practice of neuromuscular blockade during cardiac surgery.196–199 Historically, high-dose opioid techniques allowed practitioners to use high doses of long-acting nondepolarizing drugs because patients had return of motor function long before amnestic and analgesic drugs were metabolized. Current fast-track early extubation techniques and the potential association between awareness and neuromuscular blockade200 have encouraged practitioners to use less paralytic drug during CPB. Optimizing neuromuscular blockade becomes even more challenging in the face of the aforementioned pharmacokinetic and pharmacodynamic alterations due to CPB. If neuromuscular blockade is planned, careful dosing should be guided by peripheral nerve stimulation with a twitch monitor. It is important to note that muscle strength decreases substantially during hypothermia, even in the absence of muscle relaxants.54 This is likely to affect the amplitude of muscle twitches seen with peripheral nerve stimulation.

CONCLUSIONS/FUTURE DIRECTIONS

The unique pharmacokinetic and pharmacodynamic alterations associated with CPB, lack of traditional clinical signs of “adequate” anesthesia, and lack of a reliable monitor of anesthetic depth make the cardiac anesthesiologist’s job of ensuring amnesia, analgesia, and muscle relaxation during CPB difficult to impossible. Much of our knowledge is based on clinical investigations performed many years ago under much different clinical management strategies (profound hypothermia, profound hemodilution, no circuit coating) than exist today (normal to mild/moderate hypothermia, autologous priming, MECC, circuit coating). Further complicating clinical management is lack of any regulatory oversight regarding after-market modification of the CPB circuit by perfusionists or occupational health and safety concerns. Last, lack of consensus regarding how to deliver anesthetics during CPB is perhaps best highlighted by the difference in clinical practice between North American practitioners (with a strong preference for volatile anesthetics) and European practitioners (generally favoring total IV anesthesia).

Strategic areas for future research include evaluating the pharmacokinetic and pharmacodynamic alterations induced by modern CPB circuitry (smaller, coated) under near-normothermic conditions, clinical ramifications of neuroinflammation associated with CPB, and development of a reliable clinical monitor of anesthetic depth. Finally, regulatory oversight mechanisms need to be developed to standardize not only CPB circuitry but also after-market modifications.

APPENDIX

Systematic Literature Review Search Strategy

We performed a systematic review of the effects of cardiopulmonary bypass on pharmacokinetics of anesthetic drugs and unintended awareness during cardiac surgery. A list of pertinent keywords was derived, and a PubMed database search was performed for each topic, anesthetic awareness and pharmacokinetics. To assess the feasibility of this technique, we began with the topic perceived to be more focused: anesthetic awareness and cardiac surgery. Search terms were “Cardiac Surgery” OR “Cardiopulmonary Bypass” AND Awareness OR Recall OR BIS AND Anesthesia NOT “General Anesthesia” NOT Epidural NOT “General Anesthesia” NOT Spinal NOT Epilepsy (12/11/13). Results of this search were reviewed, and titles and abstracts related to the topic were highlighted. Recently published, highly cited, and review articles were considered “high-yield” articles, and the PubMed “related citations” query was applied to each. Articles related to the topic highlighted during this round were added to the list. The “related citations” query was also applied to “high-yield” articles from this round. Any related articles identified during this final round were added to the list. Articles in languages other than English were excluded from our search.

A similar search strategy was used for pharmacokinetics. The only notable difference in technique was restricting the initial screen to articles published from 2000 to the present. Search terms were “Cardiac Surgery” OR “Cardiopulmonary Bypass” OR “Open Heart” AND Pharmacokinetics OR Pharmacodynamics AND Anesthesia OR Anesthetic NOT Cardioprotection NOT Epidural NOT Spinal NOT Non-Cardiac NOT Preconditioning NOT Cardioplegia NOT Transfusion, Filters: Publication date from 2000/01/01 to 2013/12/31; Humans; English (12/17/13).

The search on awareness and cardiac surgery yielded 1349 results. Titles and abstracts related to the topic narrowed the list to 54. Seven “high-yield” articles were identified in this round.149,172,176,179,201–203 As expected, there was significant overlap in studies found using the “related citations” query. However, 35 additional articles were identified. Five articles from during this round were considered “high yield,” and the “related citations” query was applied to each.141,143,175,181,182 Fifteen additional relevant studies were discovered and added to the total. In summary, 104 articles pertinent to the topic of anesthetic awareness during cardiac surgery were identified and obtained for full review.

The same systematic approach was then turned to pharmacokinetics and CPB. The initial screen yielded 1348 results. Titles and abstracts related to the topic narrowed the list to 113. Nine “high-yield” articles were identified in this round.3,4,14,17,18,51,96,103,120 The “related citations” query yielded 99 more articles. Five more “high-yield” articles were analyzed.7,193,204–206 Sixteen additional studies were identified. In summary, 228 articles pertinent to the topic of pharmacokinetics and CPB were identified and obtained for full review. Bibliographies of all articles obtained for review were used to identify seminal papers for historical context.

The search strategy and results are depicted in Figures 11 and 12.

F11-13
Figure 11:
Systematic literature review search results for awareness and cardiac surgery.
F12-13
Figure 12:
Systematic literature review search for pharmacokinetics and cardiopulmonary bypass.

DISCLOSURES

Name: Aaron E. Barry, MD.

Contribution: This author helped design the study, conduct the study, analyze the data, and write the manuscript.

Attestation: Aaron E. Barry approved the final manuscript.

Name: Mark A. Chaney, MD.

Contribution: This author helped design the study, conduct the study, analyze the data, and write the manuscript.

Attestation: Mark A. Chaney approved the final manuscript.

Name: Martin J. London, MD.

Contribution: This author helped design the study, conduct the study, analyze the data, and write the manuscript.

Attestation: Martin J. London approved the final manuscript.

RECUSE NOTE

Dr. Martin London is the Section Editor for Perioperative Echocardiography and Cardiovascular Education for Anesthesia & Analgesia. This manuscript was handled by Dr. Steven L. Shafer, Editor-in-Chief, and Dr. London was not involved in any way with the editorial process or decision.

REFERENCES

1. Murphy GS, Hessel EA II, Groom RC. Optimal perfusion during cardiopulmonary bypass: an evidence-based approach. Anesth Analg. 2009;108:1394–417
2. Eger EI II, Sonner JM. Anaesthesia defined (gentlemen, this is no humbug). Best Pract Res Clin Anaesthesiol. 2006;20:23–9
3. Hall RI. Cardiopulmonary bypass and the systemic inflammatory response: effects on drug action. J Cardiothorac Vasc Anesth. 2002;16:83–98
4. Mets B. The pharmacokinetics of anesthetic drugs and adjuvants during cardiopulmonary bypass. Acta Anaesthesiol Scand. 2000;44:261–73
5. Gravlee GP, Davis RF, Kurusz M, Utley JR Cardiopulmonary Bypass: Principles and Practice. 20002nd ed. Philadelphia, PA Wolters Kluwer Health/Lippincott Williams & Wilkins
6. . American Society of ExtraCorporeal Technology Standards and Guidelines for Perfusion Practice 2013. Available at: http://www.amsect.org/sections/practice/practice-articles/article.iphtml. Accessed December 18, 2013
7. Nigro Neto C, Landoni G, Cassarà L, De Simone F, Zangrillo A, Tardelli MA. Use of volatile anesthetics during cardiopulmonary bypass: a systematic review of adverse events. J Cardiothorac Vasc Anesth. 2014;28:84–9
8. Hickey S, Gaylor JD, Kenny GN. In vitro uptake and elimination of isoflurane by different membrane oxygenators. J Cardiothorac Vasc Anesth. 1996;10:352–5
9. Philipp A, Wiesenack C, Behr R, Schmid FX, Birnbaum DE. High risk of intraoperative awareness during cardiopulmonary bypass with isoflurane administration via diffusion membrane oxygenators. Perfusion. 2002;17:175–8
10. Guidance for Cardiopulmonary Bypass Oxygenators 510(k) Submissions; Final Guidance for Industry and FDA Staff, 2000.
11. Nigro Neto C, Arnoni R, Rida BS, Landoni G, Tardelli MA. Randomized trial on the effect of sevoflurane on polypropylene membrane oxygenator performance. J Cardiothorac Vasc Anesth. 2013;27:903–7
12. Wiesenack C, Wiesner G, Keyl C, Gruber M, Philipp A, Ritzka M, Prasser C, Taeger K. In vivo uptake and elimination of isoflurane by different membrane oxygenators during cardiopulmonary bypass. Anesthesiology. 2002;97:133–8
13. Hiraoka H, Yamamoto K, Okano N, Morita T, Goto F, Horiuchi R. Changes in drug plasma concentrations of an extensively bound and highly extracted drug, propofol, in response to altered plasma binding. Clin Pharmacol Ther. 2004;75:324–30
14. Takizawa E, Hiraoka H, Takizawa D, Goto F. Changes in the effect of propofol in response to altered plasma protein binding during normothermic cardiopulmonary bypass. Br J Anaesth. 2006;96:179–85
15. Yoshitani K, Kawaguchi M, Takahashi M, Kitaguchi K, Furuya H. Plasma propofol concentration and EEG burst suppression ratio during normothermic cardiopulmonary bypass. Br J Anaesth. 2003;90:122–6
16. Dawson PJ, Bjorksten AR, Blake DW, Goldblatt JC. The effects of cardiopulmonary bypass on total and unbound plasma concentrations of propofol and midazolam. J Cardiothorac Vasc Anesth. 1997;11:556–61
17. Nitzschke R, Wilgusch J, Kersten JF, Trepte CJ, Haas SA, Reuter DA, Goetz AE, Goepfert MS. Changes in sevoflurane plasma concentration with delivery through the oxygenator during on-pump cardiac surgery. Br J Anaesth. 2013;110:957–65
18. Jeleazcov C, Saari TI, Ihmsen H, Schüttler J, Fechner J. Changes in total and unbound concentrations of sufentanil during target controlled infusion for cardiac surgery with cardiopulmonary bypass. Br J Anaesth. 2012;109:698–706
19. Hudson RJ, Thomson IR, Jassal R. Effects of cardiopulmonary bypass on sufentanil pharmacokinetics in patients undergoing coronary artery bypass surgery. Anesthesiology. 2004;101:862–71
20. Hudson RJ, Thomson IR, Jassal R, Peterson DJ, Brown AD, Freedman JI. Cardiopulmonary bypass has minimal effects on the pharmacokinetics of fentanyl in adults. Anesthesiology. 2003;99:847–54
21. Barvais L, Heitz D, Schmartz D, Maes V, Coussaert E, Cantraine F, d’Hollander A. Pharmacokinetic model-driven infusion of sufentanil and midazolam during cardiac surgery: assessment of the prospective predictive accuracy and the quality of anesthesia. J Cardiothorac Vasc Anesth. 2000;14:402–8
22. Michelsen LG, Holford NH, Lu W, Hoke JF, Hug CC, Bailey JM. The pharmacokinetics of remifentanil in patients undergoing coronary artery bypass grafting with cardiopulmonary bypass. Anesth Analg. 2001;93:1100–5
23. Hammarén E, Yli-Hankala A, Rosenberg PH, Hynynen M. Cardiopulmonary bypass-induced changes in plasma concentrations of propofol and in auditory evoked potentials. Br J Anaesth. 1996;77:360–4
24. Tarr TJ, Snowdon SL. Blood/gas solubility coefficient and blood concentration of enflurane during normothermic and hypothermic cardiopulmonary bypass. J Cardiothorac Vasc Anesth. 1991;5:111–5
25. Mazoit JX, Samii K. Binding of propofol to blood components: implications for pharmacokinetics and for pharmacodynamics. Br J Clin Pharmacol. 1999;47:35–42
26. Yu RG, Zhou JX, Liu J. Prediction of volatile anaesthetic solubility in blood and priming fluids for extracorporeal circulation. Br J Anaesth. 2001;86:338–44
27. Nanjappa A, Gill J, Sadat U, Colah S, Abu-Omar Y, Nair S. The effect of retrograde autologous priming on intraoperative blood product transfusion in coronary artery bypass grafting. Perfusion. 2013;28:530–5
28. Sun P, Ji B, Sun Y, Zhu X, Liu J, Long C, Zheng Z. Effects of retrograde autologous priming on blood transfusion and clinical outcomes in adults: a meta-analysis. Perfusion. 2013;28:238–43
29. Murphy GS, Szokol JW, Nitsun M, Alspach DA, Avram MJ, Vender JS, Votapka TV, Rosengart TK. The failure of retrograde autologous priming of the cardiopulmonary bypass circuit to reduce blood use after cardiac surgical procedures. Anesth Analg. 2004;98:1201–7
30. Murphy GS, Szokol JW, Nitsun M, Alspach DA, Avram MJ, Vender JS, DeMuro N, Hoff WJ. Retrograde autologous priming of the cardiopulmonary bypass circuit: safety and impact on postoperative outcomes. J Cardiothorac Vasc Anesth. 2006;20:156–61
31. Fromes Y, Gaillard D, Ponzio O, Chauffert M, Gerhardt MF, Deleuze P, Bical OM. Reduction of the inflammatory response following coronary bypass grafting with total minimal extracorporeal circulation. Eur J Cardiothorac Surg. 2002;22:527–33
32. Anastasiadis K, Asteriou C, Antonitsis P, Argiriadou H, Grosomanidis V, Kyparissa M, Deliopoulos A, Konstantinou D, Tossios P. Enhanced recovery after elective coronary revascularization surgery with minimal versus conventional extracorporeal circulation: a prospective randomized study. J Cardiothorac Vasc Anesth. 2013;27:859–64
33. Kussman BD, Zurakowski D, Sullivan L, McGowan FX, Davis PJ, Laussen PC. Evaluation of plasma fentanyl concentrations in infants during cardiopulmonary bypass with low-volume circuits. J Cardiothorac Vasc Anesth. 2005;19:316–21
34. Benet LZ, Hoener BA. Changes in plasma protein binding have little clinical relevance. Clin Pharmacol Ther. 2002;71:115–21
35. du Souich P, Verges J, Erill S. Plasma protein binding and pharmacological response. Clin Pharmacokinet. 1993;24:435–40
36. Engdahl O, Abrahams M, Björnsson A, Vegfors M, Norlander B, Ahlner J, Eintrei C. Cerebrospinal fluid concentrations of propofol during anaesthesia in humans. Br J Anaesth. 1998;81:957–9
37. Schnider TW, Minto CF, Shafer SL, Gambus PL, Andresen C, Goodale DB, Youngs EJ. The influence of age on propofol pharmacodynamics. Anesthesiology. 1999;90:1502–16
38. Takizawa D, Hiraoka H, Goto F, Yamamoto K, Horiuchi R. Human kidneys play an important role in the elimination of propofol. Anesthesiology. 2005;102:327–30
39. Hiraoka H, Yamamoto K, Miyoshi S, Morita T, Nakamura K, Kadoi Y, Kunimoto F, Horiuchi R. Kidneys contribute to the extrahepatic clearance of propofol in humans, but not lungs and brain. Br J Clin Pharmacol. 2005;60:176–82
40. Zamacona MK, Suárez E, Aguilera L, Rodríguez-Sasiaín, Aguirre C, Calvo R. Serum protein binding of propofol in critically ill patients. Acta Anaesthesiol Scand. 1997;41:1267–72
41. Gartner SL, Vahouny GV. Heparin activation of soluble heart lipoprotein lipase. Am J Physiol. 1966;211:1063–8
42. Kessler KM, Leech RC, Spann JF. Blood collection techniques, heparin and quinidine protein binding. Clin Pharmacol Ther. 1979;25:204–10
43. Storstein L, Nitter-Hauge S, Fjeld N. Effect of cardiopulmonary bypass with heparin administration on digitoxin pharmacokinetics, serum electrolytes, free fatty acids, and renal function. J Cardiovasc Pharmacol. 1979;1:191–204
44. Wood M, Shand DG, Wood AJ. Altered drug binding due to the use of indwelling heparinized cannulas (heparin lock) for sampling. Clin Pharmacol Ther. 1979;25:103–7
45. Wood M, Shand DG, Wood AJ. Propranolol binding in plasma during cardiopulmonary bypass. Anesthesiology. 1979;51:512–6
46. Wood M. Plasma drug binding: implications for anesthesiologists. Anesth Analg. 1986;65:786–804
47. Landow L, Wilson J, Heard SO, Townsend P, VanderSalm TJ, Okike ON, Pezzella TA, Pasque M. Free and total lidocaine levels in cardiac surgical patients. J Cardiothorac Anesth. 1990;4:340–7
48. Bennett-Guerrero E, Sorohan JG, Canada AT, Ayuso L, Newman MF, Reves JG, Mythen MG. epsilon-Aminocaproic acid plasma levels during cardiopulmonary bypass. Anesth Analg. 1997;85:248–51
49. Larsen JA. The effect of cooling on liver function in cats. Acta Physiol Scand. 1971;81:197–207
50. McAllister RG Jr, Tan TG. Effect of hypothermia on drug metabolism. In vitro studies with propranolol and verapamil. Pharmacology. 1980;20:95–100
51. Withington D, Ménard G, Varin F. Cisatracurium pharmacokinetics and pharmacodynamics during hypothermic cardiopulmonary bypass in infants and children. Paediatr Anaesth. 2011;21:341–6
52. Cammu G, Coddens J, Hendrickx J, Deloof T. Dose requirements of infusions of cisatracurium or rocuronium during hypothermic cardiopulmonary bypass. Br J Anaesth. 2000;84:587–90
53. Sam WJ, Hammer GB, Drover DR. Population pharmacokinetics of remifentanil in infants and children undergoing cardiac surgery. BMC Anesthesiol. 2009;9:5
54. Heier T, Caldwell JE. Impact of hypothermia on the response to neuromuscular blocking drugs. Anesthesiology. 2006;104:1070–80
55. Grigore AM, Murray CF, Ramakrishna H, Djaiani G. A core review of temperature regimens and neuroprotection during cardiopulmonary bypass: does rewarming rate matter? Anesth Analg. 2009;109:1741–51
56. Mori A, Watanabe K, Onoe M, Watarida S, Nakamura Y, Magara T, Tabata R, Okada Y. Regional blood flow in the liver, pancreas and kidney during pulsatile and nonpulsatile perfusion under profound hypothermia. Jpn Circ J. 1988;52:219–27
57. Cook DJ, Orszulak TA, Daly RC. The effects of pulsatile cardiopulmonary bypass on cerebral and renal blood flow in dogs. J Cardiothorac Vasc Anesth. 1997;11:420–7
58. Hampton WW, Townsend MC, Schirmer WJ, Haybron DM, Fry DE. Effective hepatic blood flow during cardiopulmonary bypass. Arch Surg. 1989;124:458–9
59. Chetty G, Sharpe DA, Nandi J, Butler SJ, Mitchell IM. Liver blood flow during cardiac surgery. Perfusion. 2004;19:153–6
60. Okano N, Miyoshi S, Owada R, Fujita N, Kadoi Y, Saito S, Goto F, Morita T. Impairment of hepatosplanchnic oxygenation and increase of serum hyaluronate during normothermic and mild hypothermic cardiopulmonary bypass. Anesth Analg. 2002;95:278–86
61. Hashimoto K, Sasaki T, Hachiya T, Onoguchi K, Takakura H, Oshiumi M, Takeuchi S. Superior hepatic mitochondrial oxidation-reduction state in normothermic cardiopulmonary bypass. J Thorac Cardiovasc Surg. 2001;121:1179–86
62. Yamada T, Nomoto S, Aota M, Nishimura K, Matsuda K, Ban T. Hepatic circulation during nonpulsatile cardiopulmonary bypass. ASAIO J. 1995;41:M294–7
63. Gårdebäck M, Settergren G, Brodin LA, Jorfeldt L, Galuska D, Ekberg K, Wahren J. Splanchnic blood flow and oxygen uptake during cardiopulmonary bypass. J Cardiothorac Vasc Anesth. 2002;16:308–15
64. Mathie RT, Ohri SK, Batten JJ, Peters AM, Keogh BE. Hepatic blood flow during cardiopulmonary bypass operations: the effect of temperature and pulsatility. J Thorac Cardiovasc Surg. 1997;114:292–3
65. McMurray TJ, Collier PS, Carson IW, Lyons SM, Elliott P. Propofol sedation after open heart surgery. A clinical and pharmacokinetic study. Anaesthesia. 1990;45:322–6
66. Maitre PO, Funk B, Crevoisier C, Ha HR. Pharmacokinetics of midazolam in patients recovering from cardiac surgery. Eur J Clin Pharmacol. 1989;37:161–6
67. Stanley TH. Arterial pressure and deltoid muscle gas tensions during cardiopulmonary bypass in man. Can Anaesth Soc J. 1978;25:286–90
68. Mandak J, Pojar M, Cibicek N, Lonsky V, Palicka V, Kakrdova D, Nedvidkova J, Kubicek J, Zivny P. Impact of cardiopulmonary bypass on peripheral tissue metabolism and microvascular blood flow. Perfusion. 2008;23:339–46
69. Boer F, Olofsen E, Bovill JG, Burm AG, Hak A, Geerts M, Wetselaar KE. Pulmonary uptake of sufentanil during and after constant rate infusion. Br J Anaesth. 1996;76:203–8
70. Rosen DA, Rosen KR. Elimination of drugs and toxins during cardiopulmonary bypass. J Cardiothorac Vasc Anesth. 1997;11:337–40
71. Okutani R, Philbin DM, Rosow CE, Koski G, Schneider RC. Effect of hypothermic hemodilutional cardiopulmonary bypass on plasma sufentanil and catecholamine concentrations in humans. Anesth Analg. 1988;67:667–70
72. Koren G, Crean P, Klein J, Goresky G, Villamater J, MacLeod SM. Sequestration of fentanyl by the cardiopulmonary bypass (CPBP). Eur J Clin Pharmacol. 1984;27:51–6
73. Hammarén E, Rosenberg PH, Hynynen M. Coating of extracorporeal circuit with heparin does not prevent sequestration of propofol in vitro. Br J Anaesth. 1999;82:38–40
74. Su HB, Tseng CC, Jenn CT, Chang CL, Huang JD. Changes of propofol levels in isolated cardiopulmonary bypass circuit. Acta Anaesthesiol Sin. 1996;34:17–20
75. Hynynen M, Hammarén E, Rosenberg PH. Propofol sequestration within the extracorporeal circuit. Can J Anaesth. 1994;41:583–8
76. Tarr TJ, Kent AP. Sequestration of propofol in an extracorporeal circuit. J Cardiothorac Anesth. 1989;3:75
77. Myers GJ, Voorhees C, Eke B, Johnstone R. The effect of Diprivan (propofol) on phosphorylcholine surfaces during cardiopulmonary bypass—an in vitro investigation. Perfusion. 2009;24:349–55
78. Marcoux J, Sohn N, McNair E, Rosin M, Smith G, Lim H, Mycyk T, Meng Q. Outcomes comparison of 5 coated cardiopulmonary bypass circuits versus an uncoated control group of patients undergoing cardiac surgery. Perfusion. 2009;24:307–15
79. Sohn N, Marcoux J, Mycyk T, Krahn J, Meng Q. The impact of different biocompatible coated cardiopulmonary bypass circuits on inflammatory response and oxidative stress. Perfusion. 2009;24:231–7
80. Marcoux JE, Mycyk TR. Are any biocompatible coatings capable of attenuating the deleterious effects of cardiopulmonary bypass? Perfusion. 2013;28:433–9
81. Reser D, Seifert B, Klein M, Dreizler T, Hasenclever P, Falk V, Starck C. Retrospective analysis of outcome data with regards to the use of Phisio®-, Bioline®- or Softline®-coated cardiopulmonary bypass circuits in cardiac surgery. Perfusion. 2012;27:530–4
82. Chaney MA. Corticosteroids and cardiopulmonary bypass: a review of clinical investigations. Chest. 2002;121:921–31
83. Stahl GL, Shernan SK, Smith PK, Levy JH. Complement activation and cardiac surgery: a novel target for improving outcomes. Anesth Analg. 2012;115:759–71
84. Belhaj A. Actual knowledge of systemic inflammation reaction during cardiopulmonary bypass. Recent Pat Cardiovasc Drug Discov. 2012;7:165–9
85. Kats S, Schönberger JP, Brands R, Seinen W, van Oeveren W. Endotoxin release in cardiac surgery with cardiopulmonary bypass: pathophysiology and possible therapeutic strategies. An update. Eur J Cardiothorac Surg. 2011;39:451–8
86. De Somer F. Recent advances in the comprehension and the management of perioperative systemic host response during cardiopulmonary bypass. Recent Pat Cardiovasc Drug Discov. 2012;7:180–5
87. Brekke HK, Hammersborg SM, Lundemoen S, Mongstad A, Kvalheim VL, Haugen O, Husby P. Isoflurane in contrast to propofol promotes fluid extravasation during cardiopulmonary bypass in pigs. Anesthesiology. 2013;119:861–70
88. Nordén I. The influence of anaesthetics on systemic vascular resistance during cardiopulmonary bypass. Scand J Thorac Cardiovasc Surg. 1974;8:81–7
89. Nussmeier NA, Moskowitz GJ, Weiskopf RB, Cohen NH, Fisher DM, Eger EI II. In vitro anesthetic washin and washout via bubble oxygenators: influence of anesthetic solubility and rates of carrier gas inflow and pump blood flow. Anesth Analg. 1988;67:982–7
90. Nussmeier NA, Lambert ML, Moskowitz GJ, Cohen NH, Weiskopf RB, Fisher DM, Eger EI II. Washin and washout of isoflurane administered via bubble oxygenators during hypothermic cardiopulmonary bypass. Anesthesiology. 1989;71:519–25
91. Zhou JX, Liu J. Dynamic changes in blood solubility of desflurane, isoflurane, and halothane during cardiac surgery. J Cardiothorac Vasc Anesth. 2001;15:555–9
92. Zhou J, Liu Y, Liu J. Combined effect of hypothermia and crystalloid hemodilution on the solubility of volatile anesthetics in human blood. Chin Med J (Engl). 2002;115:1014–9
93. Lerman J, Gregory GA, Eger EI II. Hematocrit and the solubility of volatile anesthetics in blood. Anesth Analg. 1984;63:911–4
94. Lockwood GG, Sapsed-Byrne SM, Smith MA. Effect of temperature on the solubility of desflurane, sevoflurane, enflurane and halothane in blood. Br J Anaesth. 1997;79:517–20
95. Eger RR, Eger EI II. Effect of temperature and age on the solubility of enflurane, halothane, isoflurane, and methoxyflurane in human blood. Anesth Analg. 1985;64:640–2
96. Goucke CR, Hackett LP, Barrett PH, Ilett KF. Blood concentrations of enflurane before, during, and after hypothermic cardiopulmonary bypass. J Cardiothorac Vasc Anesth. 2007;21:218–23
97. Mets B, Reich NT, Mellas N, Beck J, Park S. Desflurane pharmacokinetics during cardiopulmonary bypass. J Cardiothorac Vasc Anesth. 2001;15:179–82
98. Zhou JX, Liu J. The effect of temperature on solubility of volatile anesthetics in human tissues. Anesth Analg. 2001;93:234–8
99. Stern RC, Weiss CI, Steinbach JH, Evers AS. Isoflurane uptake and elimination are delayed by absorption of anesthetic by the Scimed membrane oxygenator. Anesth Analg. 1989;69:657–62
100. Henderson JM, Nathan HJ, Lalande M, Winkler MH, Dubé LM. Washin and washout of isoflurane during cardiopulmonary bypass. Can J Anaesth. 1988;35:587–90
101. Rödig G, Keyl C, Wiesner G, Philipp A, Hobbhahn J. Effects of sevoflurane and isoflurane on systemic vascular resistance: use of cardiopulmonary bypass as a study model. Br J Anaesth. 1996;76:9–12
102. Bauer M, Wilhelm W, Kraemer T, Kreuer S, Brandt A, Adams HA, Hoff G, Larsen R. Impact of bispectral index monitoring on stress response and propofol consumption in patients undergoing coronary artery bypass surgery. Anesthesiology. 2004;101:1096–104
103. Mathew PJ, Puri GD, Dhaliwal RS. Propofol requirement titrated to bispectral index: a comparison between hypothermic and normothermic cardiopulmonary bypass. Perfusion. 2009;24:27–32
104. Hayashida M, Sekiyama H, Orii R, Chinzei M, Ogawa M, Arita H, Hanaoka K, Takamoto S. Effects of deep hypothermic circulatory arrest with retrograde cerebral perfusion on electroencephalographic bispectral index and suppression ratio. J Cardiothorac Vasc Anesth. 2007;21:61–7
105. Hett DA, Smith DC, Pilkington SN, Abbott TR. Effect of temperature and cardiopulmonary bypass on the auditory evoked response. Br J Anaesth. 1995;75:293–6
106. Chandran Mahaldar DA, Gadhinglajkar S, Sreedhar R. Sevoflurane requirement to maintain bispectral index-guided steady-state level of anesthesia during the rewarming phase of cardiopulmonary bypass with moderate hypothermia. J Cardiothorac Vasc Anesth. 2013;27:59–62
107. Liu EH, Dhara SS. Monitoring oxygenator expiratory isoflurane concentrations and the bispectral index to guide isoflurane requirements during cardiopulmonary bypass. J Cardiothorac Vasc Anesth. 2005;19:485–7
108. Liu M, Hu X, Liu J. The effect of hypothermia on isoflurane MAC in children. Anesthesiology. 2001;94:429–32
109. Laussen PC, Murphy JA, Zurakowski D, Sullivan LJ, McGowan FX Jr, Demaso DR. Bispectral index monitoring in children undergoing mild hypothermic cardiopulmonary bypass. Paediatr Anaesth. 2001;11:567–73
110. Antognini JF. Hypothermia eliminates isoflurane requirements at 20 degrees C. Anesthesiology. 1993;78:1152–6
111. Schmidlin D, Hager P, Schmid ER. Monitoring level of sedation with bispectral EEG analysis: comparison between hypothermic and normothermic cardiopulmonary bypass. Br J Anaesth. 2001;86:769–76
112. Mathew JP, Weatherwax KJ, East CJ, White WD, Reves JG. Bispectral analysis during cardiopulmonary bypass: the effect of hypothermia on the hypnotic state. J Clin Anesth. 2001;13:301–5
113. Honan D, Doherty D, Frizelle H. A comparison of the effects on bispectral index of mild vs. moderate hypothermia during cardiopulmonary bypass. Eur J Anaesthesiol. 2006;23:385–90
114. Ziegeler S, Buchinger H, Wilhelm W, Larsen R, Kreuer S. Impact of deep hypothermic circulatory arrest on the BIS index. J Clin Anesth. 2010;22:340–5
115. Doi M, Gajraj RJ, Mantzaridis H, Kenny GN. Effects of cardiopulmonary bypass and hypothermia on electroencephalographic variables. Anaesthesia. 1997;52:1048–55
116. Driessen JJ, Harbers JB, van Egmond J, Booij LH. Evaluation of the electroencephalographic bispectral index during fentanyl-midazolam anaesthesia for cardiac surgery. Does it predict haemodynamic responses during endotracheal intubation and sternotomy? Eur J Anaesthesiol. 1999;16:622–7
117. Dewandre PY, Hans P, Bonhomme V, Brichant JF, Lamy M. Effects of mild hypothermic cardiopulmonary bypass on EEG bispectral index. Acta Anaesthesiol Belg. 2000;51:187–90
118. Puig MM, Warner W, Tang CK, Laorden ML, Turndorf H. Effects of temperature on the interaction of morphine with opioid receptors. Br J Anaesth. 1987;59:1459–64
119. Kansanaho M, Hynynen M, Olkkola KT. Model-driven closed-loop feedback infusion of atracurium and vecuronium during hypothermic cardiopulmonary bypass. J Cardiothorac Vasc Anesth. 1997;11:58–61
120. Withington D, Menard G, Harris J, Kulkarni P, Donati F, Varin F. Vecuronium pharmacokinetics and pharmacodynamics during hypothermic cardiopulmonary bypass in infants and children. Can J Anaesth. 2000;47:1188–95
121. Smeulers NJ, Wierda JM, van den Broek L, Gallandat Huet RC, Hennis PJ. Effects of hypothermic cardiopulmonary bypass on the pharmacodynamics and pharmacokinetics of rocuronium. J Cardiothorac Vasc Anesth. 1995;9:700–5
122. Antognini JF, Kien ND. Cardiopulmonary bypass does not alter canine enflurane requirements. Anesthesiology. 1992;76:953–7
123. Doak GJ, Li G, Hall RI, Sullivan JA. Does hypothermia or hyperventilation affect enflurane MAC reduction following partial cardiopulmonary bypass in dogs? Can J Anaesth. 1993;40:176–82
124. Hall RI, Sullivan JA. Does cardiopulmonary bypass alter enflurane requirements for anesthesia? Anesthesiology. 1990;73:249–55
125. Neumeister MW, Li G, Williams G, Doak G, Sullivan JA, Hall RI. Factors influencing MAC reduction after cardiopulmonary bypass in dogs. Can J Anaesth. 1997;44:1120–6
126. Yang H, Homi HM, Smith BE, Grocott HP. Cardiopulmonary bypass reduces the minimum alveolar concentration for isoflurane. J Cardiothorac Vasc Anesth. 2004;18:620–3
127. Lundell JC, Scuderi PE, Butterworth JF IV. Less isoflurane is required after than before cardiopulmonary bypass to maintain a constant bispectral index value. J Cardiothorac Vasc Anesth. 2001;15:551–4
128. Ahonen J, Sahlman A, Yli-Hankala A, Eriksson H, Nemlander A, Rämö J, Salmenperä M. No effect of cardiopulmonary bypass on hypnosis in patients anaesthetized with propofol and alfentanil. Br J Anaesth. 2004;92:137–9
129. Barbosa RA, Santos SR, White PF, Pereira VA, Silva Filho CR, Malbouisson LM, Carmona MJ. Effects of cardiopulmonary bypass on propofol pharmacokinetics and bispectral index during coronary surgery. Clinics (Sao Paulo). 2009;64:215–21
130. Newman MF, Kirchner JL, Phillips-Bute B, Gaver V, Grocott H, Jones RH, Mark DB, Reves JG, Blumenthal JANeurological Outcome Research Group and the Cardiothoracic Anesthesiology Research Endeavors Investigators. . Longitudinal assessment of neurocognitive function after coronary-artery bypass surgery. N Engl J Med. 2001;344:395–402
131. Stump DA, Rogers AT, Hammon JW, Newman SP. Cerebral emboli and cognitive outcome after cardiac surgery. J Cardiothorac Vasc Anesth. 1996;10:113–8
132. Harris DN, Bailey SM, Smith PL, Taylor KM, Oatridge A, Bydder GM. Brain swelling in first hour after coronary artery bypass surgery. Lancet. 1993;342:586–7
133. Vacas S, Degos V, Feng X, Maze M. The neuroinflammatory response of postoperative cognitive decline. Br Med Bull. 2013;106:161–78
134. Terrando N, Eriksson LI, Ryu JK, Yang T, Monaco C, Feldmann M, Jonsson Fagerlund M, Charo IF, Akassoglou K, Maze M. Resolving postoperative neuroinflammation and cognitive decline. Ann Neurol. 2011;70:986–95
135. van Harten AE, Scheeren TW, Absalom AR. A review of postoperative cognitive dysfunction and neuroinflammation associated with cardiac surgery and anaesthesia. Anaesthesia. 2012;67:280–93
136. Lockwood GG, Sapsed-Byrne SM, Adams S. A comparison of anaesthetic tensions in arterial blood and oxygenator exhaust gas during cardiopulmonary bypass. Anaesthesia. 1999;54:434–6
137. Bailey JM, Mora CT, Shafer SL. Pharmacokinetics of propofol in adult patients undergoing coronary revascularization. The Multicenter Study of Perioperative Ischemia Research Group. Anesthesiology. 1996;84:1288–97
138. Fiset P, Mathers L, Engstrom R, Fitzgerald D, Brand SC, Hsu F, Shafer SL. Pharmacokinetics of computer-controlled alfentanil administration in children undergoing cardiac surgery. Anesthesiology. 1995;83:944–55
139. Mashour GA, Orser BA, Avidan MS. Intraoperative awareness: from neurobiology to clinical practice. Anesthesiology. 2011;114:1218–33
140. Nielsen DV, Hansen MK, Johnsen SP, Hansen M, Hindsholm K, Jakobsen CJ. Health outcomes with and without use of inotropic therapy in cardiac surgery: results of a propensity score-matched analysis. Anesthesiology. 2014;120:1098–108
141. Myles PS, Leslie K, McNeil J, Forbes A, Chan MT. Bispectral index monitoring to prevent awareness during anaesthesia: the B-Aware randomised controlled trial. Lancet. 2004;363:1757–63
142. Avidan MS, Zhang L, Burnside BA, Finkel KJ, Searleman AC, Selvidge JA, Saager L, Turner MS, Rao S, Bottros M, Hantler C, Jacobsohn E, Evers AS. Anesthesia awareness and the bispectral index. N Engl J Med. 2008;358:1097–108
143. Avidan MS, Jacobsohn E, Glick D, Burnside BA, Zhang L, Villafranca A, Karl L, Kamal S, Torres B, O’Connor M, Evers AS, Gradwohl S, Lin N, Palanca BJ, Mashour GABAG-RECALL Research Group. . Prevention of intraoperative awareness in a high-risk surgical population. N Engl J Med. 2011;365:591–600
144. Sandin RH, Enlund G, Samuelsson P, Lennmarken C. Awareness during anaesthesia: a prospective case study. Lancet. 2000;355:707–11
145. Phillips AA, McLean RF, Devitt JH, Harrington EM. Recall of intraoperative events after general anaesthesia and cardiopulmonary bypass. Can J Anaesth. 1993;40:922–6
146. Ranta S, Jussila J, Hynynen M. Recall of awareness during cardiac anaesthesia: influence of feedback information to the anaesthesiologist. Acta Anaesthesiol Scand. 1996;40:554–60
147. Dowd NP, Cheng DC, Karski JM, Wong DT, Munro JA, Sandler AN. Intraoperative awareness in fast-track cardiac anesthesia. Anesthesiology. 1998;89:1068–73
148. Ranta SO, Herranen P, Hynynen M. Patients’ conscious recollections from cardiac anesthesia. J Cardiothorac Vasc Anesth. 2002;16:426–30
149. Avidan MS, Mashour GA. Prevention of intraoperative awareness with explicit recall: making sense of the evidence. Anesthesiology. 2013;118:449–56
150. Bailey AR, Jones JG. Patients’ memories of events during general anaesthesia. Anaesthesia. 1997;52:460–76
151. Iselin-Chaves IA, Flaishon R, Sebel PS, Howell S, Gan TJ, Sigl J, Ginsberg B, Glass PS. The effect of the interaction of propofol and alfentanil on recall, loss of consciousness, and the bispectral index. Anesth Analg. 1998;87:949–55
152. Sebel PS, Bowdle TA, Ghoneim MM, Rampil IJ, Padilla RE, Gan TJ, Domino KB. The incidence of awareness during anesthesia: a multicenter United States study. Anesth Analg. 2004;99:833–9
153. De Hert SG, Van der Linden PJ, Cromheecke S, Meeus R, Nelis A, Van Reeth V, ten Broecke PW, De Blier IG, Stockman BA, Rodrigus IE. Cardioprotective properties of sevoflurane in patients undergoing coronary surgery with cardiopulmonary bypass are related to the modalities of its administration. Anesthesiology. 2004;101:299–310
154. Pagel PS. Myocardial protection by volatile anesthetics in patients undergoing cardiac surgery: a critical review of the laboratory and clinical evidence. J Cardiothorac Vasc Anesth. 2013;27:972–82
155. Arya VK, Kumar A, Thingnam SK. Propofol infusion into the pump during cardiopulmonary bypass: is it safe and effective? J Cardiothorac Vasc Anesth. 2004;18:122–3
156. Prasser C, Zelenka M, Gruber M, Philipp A, Keyser A, Wiesenack C. Elimination of sevoflurane is reduced in plasma-tight compared to conventional membrane oxygenators. Eur J Anaesthesiol. 2008;25:152–7
157. . Council Directive 93/42/EEC, 1993. Available at: http://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=CONSLEG:1993L0042:20071011:EN:PDF. Accessed June 2, 2014
158. Sakamoto H, Mayumi T, Morimoto Y, Kemmotsu O, Wakisaka H, Ohno T. Sevoflurane metabolite production in a small cohort of coronary artery bypass graft surgery patients. J Cardiothorac Vasc Anesth. 2002;16:463–7
159. Cooper S, Levin R. Near catastrophic oxygenator failure. Anesthesiology. 1987;66:101–2
160. Walls JT, Curtis JJ, McClatchey BJ, Wood D. Adverse effects of anesthetic agents on polycarbonate plastic oxygenators. J Thorac Cardiovasc Surg. 1988;96:667–8
161. Maltry DE, Eggers GW Jr.. Isoflurane-induced failure of the Bentley-10 oxygenator. Anesthesiology. 1987;66:100–1
162. Lim HS, Cho SH, Kim DK, Lee SK, Ko SH. Isoflurane cracks the polycarbonate connector of extra-corporeal circuit—a case report. Korean J Anesthesiol. 2010;58:304–6
163. Hoerauf K, Harth M, Wild K, Hobbhahn J. Occupational exposure to desflurane and isoflurane during cardiopulmonary bypass: is the gas outlet of the membrane oxygenator an operating theatre pollution hazard? Br J Anaesth. 1997;78:378–80
164. Blokker-Veldhuis MJ, Rutten PM, De Hert SG. Occupational exposure to sevoflurane during cardiopulmonary bypass. Perfusion. 2011;26:383–9
165. . Anesthetic Gases: Guidelines for Workplace Exposures. Available at: https://www.osha.gov/dts/osta/anestheticgases/index.html. Accessed June 2, 2014
166. Martinez EA, Marsteller JA, Thompson DA, Gurses AP, Goeschel CA, Lubomski LH, Kim GR, Bauer L, Pronovost PJ. The Society of Cardiovascular Anesthesiologists’ FOCUS initiative: Locating Errors through Networked Surveillance (LENS) project vision. Anesth Analg. 2010;110:307–11
167. Martinez EA, Thompson DA, Errett NA, Kim GR, Bauer L, Lubomski LH, Gurses AP, Marsteller JA, Mohit B, Goeschel CA, Pronovost PJ. Review article: high stakes and high risk: a focused qualitative review of hazards during cardiac surgery. Anesth Analg. 2011;112:1061–74
168. Thompson DA, Marsteller JA, Pronovost PJ, Gurses A, Lubomski LH, Goeschel CA, Gosbee JW, Wahr J, Martinez EA. Locating errors through networked surveillance: a multimethod approach to peer assessment, hazard identification, and prioritization of patient safety efforts in cardiac surgery. J Patient Saf. 2014; [Epub ahead of print]
169. Spiess BD. Human error in medicine: change in cardiac operating rooms through the FOCUS initiative. J Extra Corpor Technol. 2011;43:P33–8
170. Wahr JA, Prager RL, Abernathy JH III, Martinez EA, Salas E, Seifert PC, Groom RC, Spiess BD, Searles BE, Sundt TM III, Sanchez JA, Shappell SA, Culig MH, Lazzara EH, Fitzgerald DC, Thourani VH, Eghtesady P, Ikonomidis JS, England MR, Sellke FW, Nussmeier NAAmerican Heart Association Council on Cardiovascular Surgery and Anesthesia, Council on Cardiovascular and Stroke Nursing, and Council on Quality of Care and Outcomes Research. . Patient safety in the cardiac operating room: human factors and teamwork: a scientific statement from the American Heart Association. Circulation. 2013;128:1139–69
171. Palmer G II, Abernathy JH III, Swinton G, Allison D, Greenstein J, Shappell S, Juang K, Reeves ST. Realizing improved patient care through human-centered operating room design: a human factors methodology for observing flow disruptions in the cardiothoracic operating room. Anesthesiology. 2013;119:1066–77
172. Kertai MD, Whitlock EL, Avidan MS. Brain monitoring with electroencephalography and the electroencephalogram-derived bispectral index during cardiac surgery. Anesth Analg. 2012;114:533–46
173. Zhang C, Xu L, Ma YQ, Sun YX, Li YH, Zhang L, Feng CS, Luo B, Zhao ZL, Guo JR, Jin YJ, Wu G, Yuan W, Yuan ZG, Yue Y. Bispectral index monitoring prevent awareness during total intravenous anesthesia: a prospective, randomized, double-blinded, multi-center controlled trial. Chin Med J (Engl). 2011;124:3664–9
174. Rampersad SE, Mulroy MF. A case of awareness despite an “adequate depth of anesthesia” as indicated by a bispectral index monitor. Anesth Analg. 2005;100:1363–4
175. Mashour GA, Shanks A, Tremper KK, Kheterpal S, Turner CR, Ramachandran SK, Picton P, Schueller C, Morris M, Vandervest JC, Lin N, Avidan MS. Prevention of intraoperative awareness with explicit recall in an unselected surgical population: a randomized comparative effectiveness trial. Anesthesiology. 2012;117:717–25
176. Punjasawadwong Y, Boonjeungmonkol N, Phongchiewboon A. Bispectral index for improving anaesthetic delivery and postoperative recovery. The Cochrane Database Syst Rev. 2007:CD003843
177. Villafranca A, Thomson IA, Grocott HP, Avidan MS, Kahn S, Jacobsohn E. The impact of bispectral index versus end-tidal anesthetic concentration-guided anesthesia on time to tracheal extubation in fast-track cardiac surgery. Anesth Analg. 2013;116:541–8
178. Fritz BA, Rao P, Mashour GA, Abdallah AB, Burnside BA, Jacobsohn E, Zhang L, Avidan MS. Postoperative recovery with bispectral index versus anesthetic concentration-guided protocols. Anesthesiology. 2013;118:1113–22
179. Pavlin JD, Souter KJ, Hong JY, Freund PR, Bowdle TA, Bower JO. Effects of bispectral index monitoring on recovery from surgical anesthesia in 1,580 inpatients from an academic medical center. Anesthesiology. 2005;102:566–73
180. Groesdonk HV, Pietzner J, Borger MA, Fassl J, Haentschel D, Paarmann H, Ender J. The incidence of intraoperative awareness in cardiac surgery fast-track treatment. J Cardiothorac Vasc Anesth. 2010;24:785–9
181. Leslie K, Myles PS, Forbes A, Chan MT. The effect of bispectral index monitoring on long-term survival in the B-aware trial. Anesth Analg. 2010;110:816–22
182. Kertai MD, Pal N, Palanca BJ, Lin N, Searleman SA, Zhang L, Burnside BA, Finkel KJ, Avidan MSB-Unaware Study Group. . Association of perioperative risk factors and cumulative duration of low bispectral index with intermediate-term mortality after cardiac surgery in the B-Unaware Trial. Anesthesiology. 2010;112:1116–27
183. Kertai MD, Palanca BJ, Pal N, Burnside BA, Zhang L, Sadiq F, Finkel KJ, Avidan MSB-Unaware Study Group. . Bispectral index monitoring, duration of bispectral index below 45, patient risk factors, and intermediate-term mortality after noncardiac surgery in the B-Unaware Trial. Anesthesiology. 2011;114:545–56
184. Welsby IJ, Ryan JM, Booth JV, Flanagan E, Messier RH, Borel CO. The bispectral index in the diagnosis of perioperative stroke: a case report and discussion. Anesth Analg. 2003;96:435–7
185. Murkin JM, Arango M. Near-infrared spectroscopy as an index of brain and tissue oxygenation. Br J Anaesth. 2009;103(Suppl 1):i3–13
186. Zanatta P, Messerotti Benvenuti S, Bosco E, Baldanzi F, Palomba D, Valfrè C. Multimodal brain monitoring reduces major neurologic complications in cardiac surgery. J Cardiothorac Vasc Anesth. 2011;25:1076–85
187. Radtke FM, Franck M, Lendner J, Krüger S, Wernecke KD, Spies CD. Monitoring depth of anaesthesia in a randomized trial decreases the rate of postoperative delirium but not postoperative cognitive dysfunction. Br J Anaesth. 2013;110(Suppl 1):i98–105
188. Sieber FE, Zakriya KJ, Gottschalk A, Blute MR, Lee HB, Rosenberg PB, Mears SC. Sedation depth during spinal anesthesia and the development of postoperative delirium in elderly patients undergoing hip fracture repair. Mayo Clin Proc. 2010;85:18–26
189. Pandit JJ, Cook TM. National Institute for Clinical Excellence guidance on measuring depth of anaesthesia: limitations of EEG-based technology. Br J Anaesth. 2013;110:325–8
190. . Practice advisory for intraoperative awareness and brain function monitoring: a report by the American Society of Anesthesiologists Task Force on intraoperative awareness. Anesthesiology. 2006;104:847–64
191. Depth of Anaesthesia Monitors: Bispectral Index (BIS), E-Entropy and Narcotrend-Compact M. 2012 Available at: http://www.nice.org.uk/nicemedia/live/13965/61547/61547.pdf. Accessed June 2, 2014
192. Shepherd J, Jones J, Frampton G, Bryant J, Baxter L, Cooper K. Technology Assessment Report commissioned by the NIHR HTA Programme on behalf of the National Institute for Health and Clinical Excellence—Diagnostics Assessment Report: Depth of Anaesthesia Monitoring (E-Entropy, Bispectral Index, and Narcotrend) 2012
193. Hendrickx JF, Carette R, Peyton P, De Wolf Ahttp://www.navat.org research consortium. . Monitoring anaesthetic gas concentrations in the exhaust of the cardiopulmonary bypass oxygenator. Br J Anaesth. 2014;112:173
194. Barr J, Fraser GL, Puntillo K, Ely EW, Gélinas C, Dasta JF, Davidson JE, Devlin JW, Kress JP, Joffe AM, Coursin DB, Herr DL, Tung A, Robinson BR, Fontaine DK, Ramsay MA, Riker RR, Sessler CN, Pun B, Skrobik Y, Jaeschke RAmerican College of Critical Care Medicine. . Clinical practice guidelines for the management of pain, agitation, and delirium in adult patients in the intensive care unit. Crit Care Med. 2013;41:263–306
195. Ji F, Li Z, Nguyen H, Young N, Shi P, Fleming N, Liu H. Perioperative dexmedetomidine improves outcomes of cardiac surgery. Circulation. 2013;127:1576–84
196. Hemmerling TM, Zaouter C. Neuromuscular blockade and outcome in cardiac anesthesia. Ann Card Anaesth. 2010;13:189–91
197. Cammu G. How rational is muscle relaxation during cardiac surgery? Acta Anaesthesiol Belg. 2007;58:7–14
198. Fleming N. Con: The choice of muscle relaxants is not important in cardiac surgery. J Cardiothorac Vasc Anesth. 1995;9:772–4
199. Gueret G, Rossignol B, Kiss G, Wargnier JP, Miossec A, Spielman S, Arvieux CC. Is muscle relaxant necessary for cardiac surgery? Anesth Analg. 2004;99:1330–3
200. Ghoneim MM, Block RI, Haffarnan M, Mathews MJ. Awareness during anesthesia: risk factors, causes and sequelae: a review of reported cases in the literature. Anesth Analg. 2009;108:527–35
201. Aranake A, Gradwohl S, Ben-Abdallah A, Lin N, Shanks A, Helsten DL, Glick DB, Jacobsohn E, Villafranca AJ, Evers AS, Avidan MS, Mashour GA. Increased risk of intraoperative awareness in patients with a history of awareness. Anesthesiology. 2013;119:1275–83
202. Leslie K, Myles PS, Forbes A, Chan MT, Short TG, Swallow SK. Recovery from bispectral index-guided anaesthesia in a large randomized controlled trial of patients at high risk of awareness. Anaesth Intensive Care. 2005;33:443–51
203. Palanca BJ, Mashour GA, Avidan MS. Processed electroencephalogram in depth of anesthesia monitoring. Curr Opin Anaesthesiol. 2009;22:553–9
204. Manfio JL, Santos LM, Carmona MJ, Dos Santos MB, Santos SR. Pharmacokinetic assessment of sufentanil in cardiac surgery. Drug Res (Stuttg). 2013;63:495–500
205. van Saet A, de Wildt SN, Knibbe CA, Bogers AD, Stolker RJ, Tibboel D. The effect of adult and pediatric cardiopulmonary bypass on pharmacokinetic and pharmacodynamic parameters. Curr Clin Pharmacol. 2013;8:297–318
206. Hemmerling TM, Russo G, Bracco D. Neuromuscular blockade in cardiac surgery: an update for clinicians. Ann Card Anaesth. 2008;11:80–90
© 2015 International Anesthesia Research Society