• Non ci sono risultati.

Propofol

N/A
N/A
Protected

Academic year: 2021

Condividi "Propofol"

Copied!
8
0
0

Testo completo

(1)

REVIEW ARTICLE

Rationale for Propofol Use in Cardiac Surgery

Lukasz J. Krzych, MD, PhD,* Dariusz Szurlej, MD, PhD,† and Andrzej Bochenek, MD, PhD*

P

ROPOFOL IS A COMMONLY USED intravenous anes-thetic agent. Chemically, propofol is a lipophilic, sterically hindered alkylated phenol that is a very weak acid.1,2 Pharma-cokinetic and pharmacodynamic properties make propofol a useful drug in everyday anesthesia with rapid and clear emer-gence, precise control of the level of sedation, and lack of cumulative effects even after prolonged administration.1-4 Al-though the terminal half-life of propofol is long, recovery is rapid because of the slow mobilization from the highly li-pophilic tissue compartment.1-4

The indications for propofol use include the induction and maintenance of anesthesia for most surgical procedures, and it can be extended into the postoperative setting or intensive care unit for sedation. Rapid redistribution and elimination make propofol valuable for short procedures and ambulatory sur-gery.1-4Moreover, the agent possesses antiemetic, antipruritic, and anticonvulsant properties.1-4

Propofol is also widely used in subjects with cardiac dis-ease.3,4An anesthetic drug for cardiac surgery should provide intraoperative amnesia, analgesia, and hemodynamic stability with minimal direct myocardial depression and rapid recovery; ideally, with weak inotropic support.4Pharmacokinetic prop-erties of propofol favor its use in clinical practice in cardiac surgery patients.3-5The induction of anesthesia with an opioid-benzodiazepine/etomidate combination followed by a mainte-nance infusion of propofol supplemented with an inhalation agent or opioid analgesic or both as needed are considered acceptable for patients undergoing routine cardiac surgery.3 Apart from general anesthesia, major indications for propofol use are sedation during painful procedures (eg, cardioversion), sternal wound debridement, central venous catheter insertion, and transesophageal echocardiography (Table 1).3,4

Propofol is safely administered to patients with cirrhosis and renal failure, with no impairment in its clearance.1However, in comparison with other agents, the induction dose of propofol has a relatively higher prevalence of respiratory depression, short-lived apnea, and arterial blood pressure (estimated even at

30%-40%) decrease.1-3 Although propofol has a negative im-pact on systemic blood pressure in cardiac surgery patients,6-9 clinical studies have shown that the agent, in combination with an opioid and adequate intravenous fluid supplementation, is a safe option for such procedures.5The other unpleasant effect of propofol is pain on injection, which can be dealt with either by a prior injection of a small amount of local anesthetic (eg, lidocaine) or by mixing it in the same syringe.1-4The previously mentioned effects are age, but not sex, dependent; the dose should be reduced in elderly patients and increased in chil-dren.1-5,10Obesity has no influence on the clinical duration of the effects of propofol (Table 2).2

A major concern is the propofol infusion syndrome (PRIS), which is rare but may be fatal if not identified early.11-14The review by Wysowski and Pollock11reported 36 cases of PRIS, including 15 pediatric and 21 adult patients, described in the literature from 1989 to 2005; and Kam and Cardone12found 61 patients with PRIS, 32 pediatric and 29 adult cases, described up to 2006. PRIS is characterized by metabolic acidosis, li-pemic plasma, hepatomegaly, rhabdomyolysis, and electrocar-diographic changes (eg, right bundle-branch block, acute bra-dycardia leading to asystole, and other cardiac arrhythmias), with no structural cardiac lesions or abnormalities in contrac-tility on echocardiography.1-5,12,13 Vasile et al14 suggested 2 groups of mechanisms directly related to the occurrence of PRIS, namely priming and triggering factors. The first group stems from severe critical illness (eg, multiple-organ failure) and central nervous system activation, resulting in the produc-tion of catecholamines and glucocorticosteroids.12,13 PRIS is believed to be triggered by a dose of propofol exceeding 4 mg/kg/h, time of infusion longer than 48 hours, and implemen-tation of concomitant infusions of endogenous catecholamines and steroids.12,13PRIS should be considered a potential serious adverse effect in high-risk cardiac surgery patients who often require catecholamine support and prolonged mechanical ven-tilation with long-lasting sedation.

Propofol has been suggested as a useful adjunct to cardiople-gic solutions because of its potential protective effect on the heart mediated by a decrease in ischemia-reperfusion injury in clinically relevant concentrations.15 Nevertheless, clinicians should be aware that cardiopulmonary bypass (CPB) alters its concentration by hemodilution, reduces clearance caused by changes in renal and hepatic blood flow, changes in unbound compared with bound drug, absorption by CPB apparatus, and hypothermia.6,7,15

Therefore, the authors have attempted to describe a rationale for propofol use in cardiac surgery. The aim of the article is to summarize data from the literature regarding the impact of the From the *1st Department of Cardiac Surgery and †Department of

Cardiac Anesthesia and Postoperative Intensive Care, Medical Uni-versity of Silesia, Katowice, Poland.

Address reprint requests to Lukasz J. Krzych, MD, PhD, 1st Depart-ment of Cardiac Surgery, Medical University of Silesia, 47 Ziolowa Street, 40-635 Katowice, Poland. E-mail:l.krzych@wp.pl

© 2009 Elsevier Inc. All rights reserved. 1053-0770/09/2306-0022$36.00/0 doi:10.1053/j.jvca.2009.05.001

Key words: propofol, cardiac surgery, coronary artery bypass graft surgery

(2)

agent on the heart, vessels, and blood, including the inflammatory system.

PROPOFOL AND THE HEART

Myocardial Contractility

The influence of propofol on the heart has been investigated in several studies. Its negative effect on myocardial contractil-ity and hemodynamic status has been revealed both in animal and human (in vitro) models.16-26The negative inotropic impact was found in nonfailing (nonischemic) and failing (acute isch-emic) myocardium in a dose-dependent manner, but only at concentrations higher than those typically used in clinical prac-tice.24,25A decrease in cardiac function was measured by sev-eral hemodynamic parameters, including left ventricular end-systolic pressure, end-diastolic length and end-systolic shortening,24 myocardial contractility, and a decrease in mean arterial pres-sure.25 Alterations in left ventricular preload, afterload, and regional chamber stiffness with impaired early-diastolic left ventricular filling26and wall-thickening fraction of the myocar-dium also were found (Table 3).26

Direct myocardial and coronary vascular responses to propo-fol were studied by Stowe et al.27 They found that propofol moderately depressed cardiac function and markedly attenuated autoregulation by causing coronary vasodilatation. In this ex-perimental in vitro study,27at concentrations below 10␮mol/L, no significant changes were observed; beyond 50 ␮mol/L, propofol caused progressive but differential decreases in heart rate, atrioventricular conduction time (leading to atrioventric-ular dissociation), left ventricatrioventric-ular pressure, ⫹LVdP/dTmax (change in the maximal positive derivative of left ventricular pressure), percent oxygen extraction, and myocardial oxygen consumption of isolated hearts.

The adverse effects of propofol in elderly and high-risk patients or those with impaired left ventricular ejection fraction are more pronounced in quantity compared with persons with a preserved cardiac function and lower perioperative risk.22,28 Nevertheless, no impact on the requirement for inotropic sup-port at the termination of CPB was found during propofol anesthesia,29,30and the simultaneous administration of calcium

chlorate during the induction of anesthesia may diminish the negative influence of propofol on cardiac function.31

Molecular Mechanisms

Previous observations depicted multiple possible biologic mechanisms linking propofol with cardiac depression including a decrease in sympathetic activity, vasodilatory effect, and modifications of ␣- and ␤-adrenoceptor binding and L-type calcium cardiac inhibition.32-36In an experimental study on rat papillary muscle, Zhou et al32found that propofol acted as a calcium channel blocker and had a direct impact on calcium channel proteins to diminish voltage-dependent L-type calcium current. Another explanation for propofol’s impact on the heart included the antagonism of ␤-adrenoreceptor binding and al-teration in receptor responsiveness to catecholamines in a dose-dependent (ranged from 25 to 200 ␮mol/L) and competitive manner.33Lejay et al34 showed that at a concentration of 45 ␮mol/L, the agent abolished the positive effect of ␣- but not ␤-adrenoreceptor stimulation. Supportive data were published by Sprung et al35who found that the impact of propofol was reversible with␤-adrenergic stimulation and mediated by re-duced calcium uptake into the sarcoplasmic reticulum. As far as adrenergic-receptor competition is concerned, the addition of propofol to dopamine may prevent dopamine-induced apopto-sis while maintaining positive inotropy by improving dopam-ine-mediated diastolic function after ischemia.36

Besides the above results from in vitro studies, the clinical evidence for propofol use in cardiac surgery patients is limited. The small number of in vivo studies, especially in humans, is a serious limitation. However, propofol has shown no significant negative inotropic effect in the clinically used concentra-tions.34,37

Table 1. Indications for Propofol Use in Cardiac Patients

Fast-track anesthesia; induction and maintenance

Sedation in cardiac ICU patients Sedation during transesophageal

echocardiography

Arterial/venous catheterization Sedation for cardioversion Sternal (other) wound

debridement Abbreviation: ICU, intensive care unit.

Table 2. Side Effects of Propofol

Vasodilatation with arterial blood pressure decrease

PRIS

Respiratory depression with apnea Histamine release

Pain on injection Venous thrombosis/

phlebitis Excitatory phenomena Hypertriglyceridemia

Table 3. Propofol and the Cardiovascular System

Myocardial contractility

Negative inotropic effect

2 Left ventricular loading (2 LVESP, 2 LVEDP, 2 SF, 2WT)

2 HR

Electrophysiology No impact on QTc interval

2 HR and 2 HRV (2 SA conduction) Atrial fibrillation inhibition (2 AV conduction,

His-Purkinje system inhibition) Molecular

mechanisms

2 Sympathetic activity

␣- and ␤-adrenoceptor response modification L-type calcium cardiac inhibition

Receptors’ sensitivity modulation Cardioprotective

effects

2 Oxidative stress (2 plasma inflammatory cytokine levels, 2 oxygen-free radicals, 2 lipid peroxidation, 2 chemotactic activity, 2 leukocyte infiltration) 2 Ischemia-reperfusion injury 2 Cardiac troponin I concentration

Vessels Arterial dilatation (pulmonary and peripheral) Venous dilatation (pulmonary and peripheral) Abbreviations: 2, decrease; LVESP, left ventricular end-systolic pressure; LVEDP, left ventricular end-diastolic pressure; SF, shorten-ing fraction; WT, wall thickenshorten-ing; HR, heart rate; HRV, heart rate variability; AV, atrioventricular; SA, sinus atrial; QTc, control QT in-terval.

(3)

Electrophysiology

Propofol has an impact on electrophysiologic findings in patients. Numerous studies revealed that during both the induc-tion and maintenance of total intravenous anesthesia, propofol significantly shortened the QT interval.38-43 Additional data suggested that propofol infusion may prolong the corrected QT (QTc) interval but only in subjects with a normal QTc interval, whereas it can be shortened in patients with a prolonged QTc interval at baseline.18However, the effects of the agent on the QT interval are generally too small to be clinically significant and to be identified in an intraoperative electrocardiogram.44 This is consistent with a study that showed no influence of propofol on QTc in healthy children (Table 3).45

Propofol infusion is also believed to decrease both heart rate and heart rate variability, predominantly by a reduction in cardiac parasympathetic tone.25,27,46-52Wu et al52found in an experimental study that the atrioventricular conduction interval may be lengthened by propofol in a dose-dependent and age-related manner, even at low concentrations (3 ␮mol/L). At higher concentrations, propofol significantly prolonged the atrioventricular conduction interval (10 ␮mol/L and more), slowed conduction through the atrial tissue (SA interval) and the His-Purkinje system (HV interval), decreased the sponta-neous heart rate, and prolonged the AV Wenckebach cycle length (all at concentrations of 30␮mol/L and higher).52These findings are consistent with in vivo studies of Cervigón et al53 who reported that anesthesia with propofol may slow atrial fibrillation and Owczuk et al54 who revealed a decrease in P-wave dispersion after propofol anesthesia.

Taking the previously described data into account, clinicians should be cautious of propofol’s electrophysiologic influence on patients with delayed or blocked conduction in the heart. Propofol can be a reasonable choice for cardiac surgery patients with atrial fibrillation, but it should be used with caution during cardioversion because of hypotension and an increase in energy required for successful effect of the procedure.3

PROPOFOL AND THE INFLAMMATORY SYSTEM

Cardiopulmonary bypass is one of the acknowledged causes of a complex systemic inflammatory response during cardiac surgery, and it may contribute to postoperative complications and even multiple-organ dysfunction.55-57 Inflammation, as a result of cellular oxidative stress and myocardial injury, is mediated by several agents including interleukins 6, 8, and 10; high-sensitivity C-reactive protein; tumor necrosis factor ␣; and the release of other inflammatory agents.55-57Thus, results suggesting that propofol has antioxidative properties58-64 and may be of great importance because of its ability to reduce ischemia-reperfusion injury in coronary artery bypass graft (CABG) surgery patients.

In an experimental study, propofol anesthesia was directly related to a significant decrease in oxidative activity measured by malondialdehyde levels in plasma and pulmonary lavage.58 In a study by Corcoran et al,59it was found that in patients undergoing CABG surgery, clinically relevant concentrations of propofol attenuated free radical-mediated and inflammatory components of myocardial reperfusion injury including modu-lation of serum concentration of malondialdehyde; systemic

concentrations of interleukins 4, 6, 8, and 10; and systemic leukocyte functions. In several investigations,60-63an antioxi-dative effect of propofol was shown via mechanisms involving decreases in neutrophil infiltration, plasma inflammatory cyto-kine levels, oxygen free-radicals production, and lipid peroxi-dation. In vitro studies also showed that propofol depresses the immunologic reaction to bacterial challenge as well as chemo-tactic activity.5

There are some supplementary data indicating that propofol may also protect erythrocytes against both oxidative and phys-ical stress, suggesting its efficacy and usefulness as an antiox-idant.64 In the latter study,64propofol increased erythrocytes’ membrane fluidity, thereby increasing their resistance to phys-ical and hemodynamic stress. Moreover, the protective effect of propofol on oxidation in red blood cell membranes was en-hanced by another antioxidant, ascorbic acid.64

Hypertriglyceridemia is common in propofol-treated pa-tients,1,3,4,65,66 and may be associated with the PRIS occur-rence.11-13In a study by Oztekin et al,67propofol was found to increase triglycerides and very low-density lipoprotein concen-trations, but not glucose concentrations a few hours after CABG surgery. Repeated measures of the lipid profile could become a valuable marker for monitoring this side effect.

CARDIOPROTECTIVE EFFECTS OF PROPOFOL

Propofol is believed to possess a cardioprotective effect that derives in part from its antioxidant properties and free radical scavenging properties.68 These properties were described in several studies.69-74Zou et al69and Xia et al70reported the role of propofol in the reduction of cardiac troponin I concentration in clinical investigations. Myocardial protection by the agent may involve the activation of protein kinase C.74-77It is sug-gested that propofol increases the sensitivity of myofibrillar actomyosin ATPase to calcium by increasing intracellular pH via the protein kinase C– dependent activation of Na⫹-H⫹ ex-change.75,76In diabetic cardiomyocytes, propofol was found to decrease myofilament Ca2⫹ sensitivity via a protein kinase C–nitric oxide synthase– dependent pathway.77Supplementary data revealed that infusion of the agent significantly reduced the number of in vitro apoptotic cells71or prevented dopamine-induced apoptosis of the heart cells after ischemia in an animal model.37

On the basis of registry data from 10,535 surgical procedures comparing the influence of sevoflurane and propofol anesthesia on clinical outcomes, Jakobsen et al concluded that propofol anesthesia may be superior in patients with severe ischemia, cardiovascular instability, or in acute/urgent surgery because of its cardioprotective properties.73 The 30-day mortality was comparable among all patients from the sevoflurane and propo-fol groups (2.84% v 3.3%, p⫽ 0.18) as well as in a subgroup of subjects with unstable angina (5.48% v 4.8%, p⫽ 0.58) or recent myocardial infarction (4.91% v 4.72%, p ⫽ 0.93). Sevoflurane anesthesia resulted in a decrease in mortality in patients with stable angina and no history of myocardial infarc-tion compared with the propofol group (2.28% v 3.14%, p⫽ 0.01).78In the cited study,78propofol anesthesia had its most important beneficial influence on 30-day mortality in urgent CABG procedures (16.23 v 8.19%, p⫽ 0.03).

(4)

outcome advantage of propofol in a multicenter prospective randomized study in 150 CABG surgery patients by Tritapepe et al.79 The postoperative concentration of troponin I was 2 times higher in anesthetized subjects from the propofol group (median⫽ 5.5 ng/dL) compared with patients from the desflu-rane group (median⫽ 2.5 ng/dL).79Moreover, patients receiv-ing a volatile anesthetic had a reduced need for postoperative inotropic support (32% v 41.3% in the propofol group, p ⫽ 0.04). Consistent findings relating to differences between propofol and sevoflurane anesthesia were shown by De Hert et al17 in a group of 20 coronary artery surgery patients. They reported that for the sevoflurane group, troponin I concentra-tions remained below the cutoff value of 2 ng/mL throughout the 36-hour observation period, but, in the propofol group, an elevation in troponin I concentrations was found 12 hours after CPB, with a peak at 24 hours followed by a decrease in the next 12 hours.17 Kohro et al74 found that the administration of propofol may also hamper previously reported cardioprotective effects of inhalation anesthetics (eg, isoflurane and sevoflurane) as well.

PROPOFOL AND THE BRAIN

Neurologic deterioration after CPB including complications of stroke, transient ischemic attacks or cognitive dysfunction is frequent.80The results of longitudinal assessment of neurocog-nitive function after CABG surgery revealed that the incidence of cognitive decline occurred in more than half the patients (53%) at discharge and remained after 5 years (46%).80 More-over, cognitive function at discharge was a major predictor of cognitive decline at 5 years.80The etiology of neuropsycho-logic deterioration is complex and includes microemboliza-tion, altered cerebral perfusion, temperature perturbations, cerebral edema, blood-brain barrier dysfunction, and a gen-eralized or localized inflammatory response.81

Proper perioperative management is required to reduce neu-rologic decline. The current literature describes several poten-tial neuroprotective interventions including perioperative propofol administration.81It has been suggested that the neu-roprotective effect of propofol is attributed to its global and regional (at brain level) antioxidative properties that were men-tioned earlier;68however, its role is believed to be beneficial only in limiting the size of cerebral damage rather than the incidence of infarction.3,81 The protective role of the de-creased cerebral mean arterial pressure caused by vasodila-tation has been neglected.81Propofol’s protective effect may be mediated by microembolic delivery reduction by way of decreasing cerebral blood flow,81 but this has not yet been confirmed in clinical trials (Table 4).

The decrease in cerebral metabolic rate and oxygen con-sumption during propofol infusion on CPB may potentially be of some importance. It was revealed that in patients undergoing CABG surgery, induction with propofol resulted in a 51% decrease in cerebral blood flow, a 36% reduction of cerebral metabolic rate and oxygen consumption, and a 25% decrease in cerebral perfusion pressure.82-84 Newman et al85 described a significant reduction in cerebral blood flow, cerebral oxygen delivery, and cerebral metabolic rate in response to propofol anesthesia during nonpulsatile normothermic and hypothermic phases of CPB. Burst-suppression doses of propofol during

moderate hypothermic CPB were also found to decrease cere-bral blood flow by 35% and cerecere-bral oxygen consumption by 10%.86

Roach et al,87however, reported no association between a reduction in cerebral metabolic suppression or a reduction in cerebral blood flow and neurologic or neuropsychologic dys-function after propofol anesthesia during CPB. By using a battery of neurologic and neuropsychologic tests in 225 pa-tients, they showed that, compared with a sufentanil group, patients in the propofol-sufentanil group tended to have an even higher incidence of adverse neurologic outcomes on postoper-ative days 1 and 2 (40% v 25%, p⫽ 0.06) and at 5 to 7 days (18% v 8%, p⫽ 0.07). These differences resolved after 50 to 70 days (6.2% v 6.2%, p⫽ 0.8).87The incidence of neuropsy-chologic deficits was comparable between the propofol and sufentanil groups (91% v 92%, p⫽ 0.73 at days 5-7, and 52%

v 47% at days 50-70, p⫽ 0.58, respectively), and neither the

severity nor the prevalence or neuropsychologic outcomes dif-fered among patients.87

In view of the previously described information, the clinical relevance of propofol use to prevent cognitive deterioration in cardiac surgery needs to be confirmed in larger prospective studies. The lack of neuroprotection in some studies may be caused by the fact that either propofol has no neuroprotective effects or that appropriate perioperative treatment diminishes any protective impact of the agent significantly (eg, proper oxygenation, glucose control, arterial filters and membrane oxygenators, and concomitant volatile anesthesia).3,81,85,87 Moreover, the methodology of the cited studies varies signifi-cantly and presents a serious limitation to reaching a conclu-sion.

VASCULAR EFFECTS OF PROPOFOL

From a cardiac surgeon’s point of view, the influence of propofol on the biologic tissues used for vascular grafts (CABG) and vasoregulation is crucial. Early postoperative graft failure attributed to spontaneous contraction or spasm requires improvements in preventative strategies and effective treatment. Thus, the potential vasodilating effect of propo-fol88-97may be crucial during the perioperative period. Unfor-tunately, there is still a discrepancy between the number of animal investigations and clinical studies concerning the influ-ence of propofol on systemic vasoregulation.88-97

Animal Studies

The well-known hypotensive effect of propofol may be mediated via multiple mechanisms including its action on pe-ripheral vasculature (arterial and venous vasodilatation) as well as a decrease in myocardial contractility, resetting of baroreflex

Table 4. Propofol and the Brain

EEG burst suppression 2 Oxidative stress 2 Cerebral metabolic rate

and oxygen consumption

2 Cerebral blood flow and cerebral blood flow velocity

2 Cerebral perfusion pressure

Microembolic delivery reduction

(5)

activity, and inhibition of the sympathetic nervous system outflow.5,19,49,88-90 The primary cause of reduction in blood pressure differs between individual studies.88-91

Data from the literature raise questions about the molecular background of vasodilatation caused by propofol, and information published about this issue is inconsistent. A number of articles confirm that vasodilatation is endothelium-dependent and uses the activation of numerous endothelial agents.88,90,95,98However, some contradictory data revealed that the effect is mediated in an endo-thelium-independent manner.92,93,99It was also found that propofol may cause significant vasodilatation in both endothelium-intact and endothelium-denuded tissue.89,100,101Thus, further investiga-tions focused on potential mechanisms of the anesthetic are needed.

Effects of the agent on arterial and venous tissue showed the changes to be dose dependent. In a study by Stowe et al,27 propofol, administered at the highest concentration (100␮mol/L), increased coronary flow by 57%⫾ 10%, decreased myocardial oxygen consumption by 37%⫾ 5%, and increased oxygen deliv-ery/myocardial oxygen consumption ratio by 150%⫾ 15%. Be-tween concentrations of 100␮mol/L and 1 mmol/L, coronary flow was maximally increased and myocardial oxygen consumption was maximally decreased by the agent. In endothelium-intact arterial rings precontracted with PGF2a, propofol did not change vascular smooth muscle tone in low concentrations (1␮mol/L-10 ␮mol/L), but at high concentrations (100 ␮mol/L-100 mmol/L) it produced a significant relaxation compared to endothelium-de-nuded rings.90

There is some evidence suggesting a quantitative role of vascular diameter during the process of relaxation.91Coughlan et al investigated the influence of propofol in canine coronary arteries and showed that small arteries showed greater vasodi-latation (at similar concentrations) than larger arteries. Vaso-dilating properties of propofol were more pronounced in distal than in proximal vessels.91

Human Studies

The impact of propofol on the vessels seems comparable in animals and humans, but the issue of the underlying mechanism is still controversial. In a study by Klockgether-Radke et al,92 propofol administered at high concentrations (100 ␮g/mL) caused an endothelium-independent relaxation on porcine and human coronary artery segments to a similar extent (⫺32% up to⫺49% and ⫺11% to ⫺67%, respectively). Wallestedt et al94

investigated the relaxant effects of propofol on smooth muscle tonus in human omental arteries and veins, and found that propofol induced relaxation of both vessels in an endothelium-independent manner. Contradictory data showed that propofol-related relaxation may be endothelium-dependent in omental and radial arteries and omental veins,95,97and Moreno et al96 found that the vasodilatation after propofol administration was similar in intact and denuded endothelium mesenteric rings. Pulmonary Vasoregulation

Park et al89 showed that propofol also promoted marked vasodilatation in pulmonary arteries. In the endothelium-intact aortic and pulmonary artery rings, the initial vasodilatation at the propofol concentration between 30 and 100␮mol/L showed gradual and partial recovery over 15 minutes, and, at a con-centration of 300␮mol/L, it caused sustained relaxation. More-over, endothelium-denuded rings and L-NAME pretreated endothelium-intact rings showed constant and sustained vaso-dilatation with all propofol concentrations.89The propofol ve-hicle had no dilating effect on vascular rings and indomethacin pretreatment decreased relaxation, the latter suggesting a me-diating role of endothelium in the process.89 Similar findings relating to the underlying mechanism were reported on a ca-nine102and rat model.103Moreover, the vasorelaxing response to propofol was seen in extrapulmonary but not in intrapulmo-nary arteries.103 Contradictory information was revealed by Horibe et al,104who found that propofol caused dose-dependent and endothelium-independent pulmonary vasorelaxation.

CONCLUSIONS

In summary, for cardiac anesthesia, propofol is useful for the induction and maintenance of anesthesia, and sedation as well, mainly because of its favorable pharmacokinetic properties. How-ever, the benefits of its use are still theoretic, and outcome advan-tages in cardiac surgical patients are unconvincing. Clinicians should be aware of serious adverse effects of propofol including hemodynamic instability, respiratory depression, and PRIS. Al-though propofol seems to be a promising vasodilator for the treatment of perioperative spasm of coronary artery grafts, there is a paucity of epidemiologic data regarding cardiac and neuropro-tective effects of the agent. Further investigations are required to explore the molecular mechanisms of propofol more precisely and to extend indications for its use in cardiac anesthetic practice.

REFERENCES 1. Vanlersberghe C, Camu F: Propofol. Handb Exp Pharmacol 182:

227-252, 2008

2. Trapani G, Altomare C, Liso G, et al: Propofol in anesthesia. Mechanism of action, structure-activity relationship, and drug delivery. Curr Med Chem 7:249-271, 2000

3. Smith I, White PF, Nathanson M, et al: Propofol. An update on its clinical use. Anesthesiology 81:1005-1043, 1994

4. Yao FSF, Malhorta V, Fontes ML (eds): Anesthesiology. Prob-lem-Oriented Patient Management (ed 6). New York, NY, Lippincott Williams and Wilkins, 2007

5. Searle NR, Sahab P: Propofol in patients with cardiac disease. Can J Anaesth 40:730-747, 1993

6. Vermeyen KM, Erpels FA, Janssen LA, et al: Propofol-fentanyl anaesthesia for coronary bypass surgery in patients with good left ventricular function. Br J Anaesth 59:1115-120, 1987

7. Vermeyen KM, De Hert SG, Erpels FA, et al: Myocardial me-tabolism during anaesthesia with propofol—Low dose fentanyl for coronary artery bypass surgery. Br J Anaesth 66:504-508, 1991

8. Russell D, Wilkes MP, Hunter SC, et al: Manual compared with target-controlled infusion of propofol. Br J Anaesth 75:562-566, 1995 9. Stephan H, Sonntag H, Schenk HD, et al: Effects of propofol on cardiovascular dynamics, myocardial blood flow and myocardial me-tabolism in patients with coronary artery disease. Br J Anaesth 58:969-975, 1986

(6)

10. Ouattara A, Boccara G, Lemaire S, et al: Target-controlled infusion of propofol and remifentanil in cardiac anaesthesia: Influence of age on predicted effect-site concentrations. Br J Anaesth 90:617-622, 2003

11. Wysowski DK, Pollock ML: Reports of death with use of propofol (Diprivan) for nonprocedural (long-term) sedation and litera-ture review. Anesthesiology 105:1047-1051, 2006

12. Kam PC, Cardone D: Propofol infusion syndrome. Anaesthesia 62:690-701, 2007

13. Rajda C, Dereczyk D, Kunkel P: Propofol infusion syndrome. J Trauma Nurs 15:118-122, 2008

14. Vasile B, Rasulo F, Candiani A, et al: The pathophysiology of propofol infusion syndrome: A simple name for a complex syndrome. Intensive Care Med 29:1417-25, 2003

15. Lim KH, Halestrap AP, Angelini GD, et al: Propofol is cardio-protective in a clinically relevant model of normothermic blood car-dioplegic arrest and cardiopulmonary bypass. Exp Biol Med (May-wood) 230:413-420, 2005

16. Ozaki M: The effects of propofol and midazolam on canine left ventricular contractility. Masui 51:611-619, 2002

17. De Hert SG, ten Broecke PW, Mertens E, et al: Sevoflurane but not propofol preserves myocardial function in coronary surgery pa-tients. Anesthesiology 97:42-49, 2002

18. Hettrick DA, Pagel PS, Warltier DC: Alterations in canine left ventricular-arterial coupling and mechanical efficiency produced by propofol. Anesthesiology 86:1088-1093, 1997

19. Filipovic M, Wang J, Michaux I, et al: Effects of halothane, sevoflurane and propofol on left ventricular diastolic function in hu-mans during spontaneous and mechanical ventilation. Br J Anaesth 94:186-192, 2005

20. Kehl F, Kress TT, Mraovic B, et al: Propofol alters left atrial function evaluated with pressure-volume relations in vivo. Anesth Analg 94:1421-1426, 2002

21. Coetzee A, Fourie P, Coetzee J, et al: Effect of various propofol plasma concentrations on regional myocardial contractility and left ventricular afterload. Anesth Analg 69:473-483, 1989

22. Sherry KM, Sartain J, Bell JH, et al: Comparison of the use of a propofol infusion in cardiac surgical patients with normal and low cardiac output states. J Cardiothorac Vasc Anesth 9:368-372, 1995

23. Hebbar L, Dorman BH, Roy RC, et al: The direct effects of propofol on myocyte contractile function after hypothermic cardiople-gic arrest. Anesth Analg 83:949-957, 1996

24. De Hert SG: Study on the effects of six intravenous anesthetic agents on regional ventricular function in dogs (thiopental, etomidate, propofol, fentanyl, sufentanil, alfentanil). Acta Anaesthesiol Belg 42: 3-39, 1999

25. Royse CF, Liew DF, Wright CE, et al: Persistent depression of contractility and vasodilation with propofol but not with sevoflurane or desflurane in rabbits. Anesthesiology 108:87-93, 2008

26. Meissner A, Weber TP, Van Aken H, et al: Recovery from myocardial stunning is faster with desflurane compared with propofol in chronically instrumented dogs. Anesth Analg 91:1333-1338, 2000

27. Stowe DF, Bosnjak ZJ, Kampine JP: Comparison of etomidate, ketamine, midazolam, propofol, and thiopental on function and metab-olism of isolated hearts. Anesth Analg 74:547-558, 1992

28. De Hert SG, Cromheecke S, ten Broecke PW, et al: Effects of propofol, desflurane, and sevoflurane on recovery of myocardial func-tion after coronary surgery in elderly high-risk patients. Anesthesiology 99:314-323, 2003

29. Hall RI, Murphy JT, Landymore R, et al: Myocardial metabolic and hemodynamic changes during propofol anesthesia for cardiac surgery in patients with reduced ventricular function. Anesth Analg 77:680-689, 1993

30. Hall RI, Murphy JT, Moffitt EA, et al: A comparison of the myocardial metabolic and haemodynamic changes produced by propo-fol-sufentanil and enflurane-sufentanil anaesthesia for patients having coronary artery bypass graft surgery. Can J Anaesth 38:996-1004, 1991 31. Tritapepe L, Voci P, Marino P, et al: Calcium chloride mini-mizes the hemodynamic effects of propofol in patients undergoing coronary artery bypass grafting. J Cardiothorac Vasc Anesth 13:150-153, 1999

32. Zhou W, Fontenot HJ, Liu S, et al: Modulation of cardiac calcium channels by propofol. Anesthesiology 86:670-675, 1997

33. Zhou W, Fontenot HJ, Wang SN, et al: Propofol-induced alter-ations in myocardial beta-adrenoceptor binding and responsiveness. Anesth Analg 89:604-608, 1999

34. Lejay M, Hanouz JL, Lecarpentier Y, et al: Modifications of the inotropic responses to alpha- and beta-adrenoceptor stimulation by propofol in rat myocardium. Anesth Analg 87:277-283, 1998

35. Sprung J, Ogletree-Hughes ML, McConnell BK, et al: The effects of propofol on the contractility of failing and nonfailing human heart muscle. Anesth Analg 93:550-559, 2001

36. Roy N, Friehs I, Cowan DB, et al: Dopamine induces postisch-emic cardiomyocyte apoptosis in vivo: an effect ameliorated by propo-fol. Ann Thorac Surg 82:2192-2199, 2006

37. Gelissen HP, Epema AH, Henning RH, et al: Inotropic effects of propofol, thiopental, midazolam, etomidate, and ketamine on isolated human atrial muscle. Anesthesiology 84:397-403, 1996

38. Paventi S, Santevecchi A, Ranieri R: Effects of sevoflurane versus propofol on QT interval. Minerva Anestesiol 67:637-640, 2001 39. Saarnivaara L, Klemola UM, Lindgren L, et al: QT interval of the ECG, heart rate and arterial pressure using propofol, methohexital or midazolam for induction of anaesthesia. Acta Anaesthesiol Scand 34:276-281, 1990

40. Saarnivaara L, Hiller A, Oikkonen M: QT interval, heart rate and arterial pressures using propofol, thiopentone or methohexitone for induction of anaesthesia in children. Acta Anaesthesiol Scand 37:419-423, 1993

41. Dennis SG, Wotton PR, Boswood A, et al: Comparison of the effects of thiopentone and propofol on the electrocardiogram of dogs. Vet Rec 160:681-686, 2007

42. Kleinsasser A, Kuenszberg E, Loeckinger A, et al: Sevoflurane, but not propofol, significantly prolongs the Q-T interval. Anesth Analg 90:25-27, 2000

43. Kanaya N, Hirata N, Kurosawa S, et al: Differential effects of propofol and sevoflurane on heart rate variability. Anesthesiology 98:34-40, 2003

44. Kim DH, Kweon TD, Nam SB, et al: Effects of target concen-tration infusion of propofol and tracheal intubation on QTc interval. Anaesthesia 63:1061-1064, 2008

45. Hume-Smith HV, Sanatani S, Lim J, et al: The effect of propofol concentration on dispersion of myocardial repolarization in children. Anesth Analg 107:806-810, 2008

46. Win NN, Kohase H, Yoshikawa F, et al: Haemodynamic changes and heart rate variability during midazolam-propofol co-in-duction. Anaesthesia 62:561-568, 2007

47. Deutschman CS, Harris AP, Fleisher LA: Changes in heart rate variability under propofol anesthesia: A possible explanation for propo-fol-induced bradycardia. Anesth Analg 79:373-377, 1994

48. Lischke V, Probst S, Behne M, et al: ST segment changes in the ECG. Anesthesia induction with propofol, etomidate or midazolam in patients with coronary heart disease. Anaesthesist 42:435-440, 1993

49. Pereira GG, Larsson MH, Yamaki FL, et al: Effects of propofol on the electrocardiogram and systolic blood pressure of healthy cats pre-medicated with acepromazine. Vet Anaesth Analg 31:235-238, 2004

(7)

50. Win NN, Fukayama H, Kohase H, et al: The different effects of intravenous propofol and midazolam sedation on hemodynamic and heart rate variability. Anesth Analg 101:97-102, 2005

51. Mäenpää M, Penttilä J, Laitio T, et al: The effects of surgical levels of sevoflurane and propofol anaesthesia on heart rate variability. Eur J Anaesthesiol 24:626-633, 2007

52. Wu MH, Su MJ, Sun SS: Age-related propofol effects on elec-trophysiological properties of isolated hearts. Anesth Analg 84:964-971, 1997

53. Cervigón R, Moreno J, Castells F, et al: Anesthesia with propo-fol slows atrial fibrillation dominant frequencies. Comput Biol Med 38:792-798, 2008

54. Owczuk R, Wujtewicz MA, Sawicka W, et al: Effect of anaes-thetic agents on p-wave dispersion on the electrocardiogram: Compar-ison of propofol and desflurane. Clin Exp Pharmacol Physiol 35:1071-1076, 2008

55. Orhan G, Sargin M, Senay S, et al: Systemic and myocardial inflammation in traditional and off-pump cardiac surgery. Tex Heart Inst J 34:160-165, 2007

56. Parolari A, Camera M, Alamanni F, et al: Systemic inflamma-tion after on-pump and off-pump coronary bypass surgery: A one-month follow-up. Ann Thorac Surg 84:823-828, 2007

57. Nesher N, Frolkis I, Vardi M, et al: Higher levels of serum cytokines and myocardial tissue markers during on-pump versus off-pump coronary artery bypass surgery. J Card Surg 21:395-402, 2006

58. Allaouchiche B, Debon R, Goudable J, et al: Oxidative stress status during exposure to propofol, sevoflurane and desflurane. Anesth Analg 93:981-985, 2001

59. Corcoran TB, Engel A, Sakamoto H, et al: The effects of propofol on lipid peroxidation and inflammatory response in elective coronary artery bypass grafting. J Cardiothorac Vasc Anesth 18:592-604, 2004

60. Rodríguez-López JM, Sánchez-Conde P, Lozano FS, et al: Lab-oratory investigation: Effects of propofol on the systemic inflammatory response during aortic surgery. Can J Anaesth 53:701-710, 2006

61. Corcoran TB, Engel A, Sakamoto H, et al: The effects of propofol on neutrophil function, lipid peroxidation and inflammatory response during elective coronary artery bypass grafting in patients with impaired ventricular function. Br J Anaesth 6;97:825-831, 2006

62. McDermott BJ, McWilliams S, Smyth K, et al: Protection of cardiomyocyte function by propofol during simulated ischemia is as-sociated with a direct action to reduce pro-oxidant activity. J Mol Cell Cardiol 42:600-608, 2007

63. Yun JY, Park KS, Kim JH, et al: Propofol reverses oxidative stress-attenuated glutamate transporter EAAT3 activity: Evidence of protein kinase C involvement. Eur J Pharmacol 565:83-88, 2007

64. Tsuchiya M, Asada A, Kasahara E, et al: Antioxidant protection of propofol and its recycling in erythrocyte membranes. Am J Respir Crit Care Med 165:54-60, 2002

65. Carrasco G, Molina R, Costa J, et al: Propofol vs midazolam in short-, medium-, and long-term sedation of critically ill patients. A cost-benefit analysis. Chest 103:557-564, 1993

66. Devlin JW, Lau AK, Tanios MA: Propofol-associated hypertri-glyceridemia and pancreatitis in the intensive care unit: An analysis of frequency and risk factors. Pharmacotherapy 25:1348-1352, 2005

67. Oztekin I, Gökdog˘an S, Oztekin DS, et al: Effects of propofol and midazolam on lipids, glucose, and plasma osmolality during and in the early postoperative period following coronary artery bypass graft surgery: A randomized trial. Yakugaku Zasshi 127:173-182, 2007

68. Vanlersberghe C, Camu F: Propofol. Handb Exp Pharmacol 182:227-252, 2008

69. Zou HD, Zhou QS, Xia WF, et al: Cardioprotective effects of propofol and midazolam in children with congenital heart diseases

undergoing open heart surgery. Zhonghua Yi Xue Za Zhi 87:2309-2312, 2007

70. Xia Z, Huang Z, Ansley DM: Large-dose propofol during car-diopulmonary bypass decreases biochemical markers of myocardial injury in coronary surgery patients: A comparison with isoflurane. Anesth Analg 103:527-532, 2006

71. Choi SU, Lee HW, Lim HJ, et al: The effects of propofol on cardiac function after 4 hours of cold cardioplegia and reperfusion. J Cardiothorac Vasc Anesth 21:678-682, 2007

72. Shao H, Li J, Zhou Y, et al: Dose-dependent protective effect of propofol against mitochondrial dysfunction in ischaemic/reperfused rat heart: role of cardiolipin. Br J Pharmacol 153:1641-1649, 2008

73. Jakobsen CJ, Berg H, Hindsholm KB, et al: The influence of propofol versus sevoflurane anesthesia on outcome in 10,535 cardiac surgical procedures. J Cardiothorac Vasc Anesth 21:664-671, 2007

74. Kohro S, Hogn QH, Nakae Y, et al: Anesthetic effects on mitochondrial ATP-sensitive K channel. Anesthesiology 95:1435-1440, 2001

75. Kanaya N, Murray PA, Damron DS: Propofol increases myo-filament Ca2⫹ sensitivity and intracellular pH via activation of Na⫹-H⫹ exchange in rat ventricular myocytes. Anesthesiology 94: 1096-1104, 2001

76. Wickley PJ, Ding X, Murray PA, et al: Propofol-induced acti-vation of protein kinase C isoforms in adult rat ventricular myocytes. Anesthesiology 104:970-977, 2006

77. Wickley PJ, Shiga T, Murray PA, et al: Propofol decreases myofilament Ca2⫹ sensitivity via a protein kinase C-, nitric oxide synthase-dependent pathway in diabetic cardiomyocytes. Anesthesiol-ogy 104:978-987, 2006

78. Gülçin I, Alici HA, Cesur M: Determination of in vitro antiox-idant and radical scavenging activities of propofol. Chem Pharm Bull (Tokyo) 53:281-285, 2005

79. Tritapepe L, Landoni G, Guarracino F, et al: Cardiac protection by volatile anaesthetics: A multicentre randomized controlled study in patients undergoing coronary artery bypass grafting wth cardiopulmo-nary bypass. Eur J Anaesthesiol 24:231-232, 2007

80. Newman MF, Kirchner JL, Phillips-Bute B, et al: Longitudinal assessment of neurocognitive function after coronary-artery bypass surgery. N Engl J Med 344:395-402, 2001

81. Grocott HP, Homi HM, Puskas F: Cognitive dysfunction after cardiac surgery: Revisiting etiology. Semin Cardiothorac Vasc Anesth 9:123-129, 2005

82. Kotani Y, Shimazawa M, Yoshimura S, et al: The experimental and clinical pharmacology of propofol, an anesthetic agent with neu-roprotective properties. CNS Neurosci Ther 14:95-106, 2008

83. Head BP, Patel P: Anesthetics and brain protection. Curr Opin Anaesthesiol 20:395-399, 2007

84. Demirci A, Unver S, Karadeniz U, et al: Middle cerebral arterial blood flow velocity and hemodynamics in heart surgery. Asian Car-diovasc Thorac Ann 15:97-101, 2007

85. Newman MF, Murkin JM, Roach G, et al: Cerebral physiologic effects of burst suppression doses of propofol during nonpulsatile cardiopulmonary bypass. CNS Subgroup of McSPI. Anesth Analg 81:452-457, 1995

86. Ederberg S, Westerlind A, Houltz E, et al: The effects of propofol on cerebral blood flow velocity and cerebral oxygen extraction during cardiopulmonary bypass. Anesth Analg 86:1201-1206, 1998

87. Roach GW, Newman MF, Murkin JM, et al: Ineffectiveness of burst suppression therapy in mitigating perioperative cerebrovascular dysfunction. Multicenter Study of Perioperative Ischemia (McSPI) Research Group. Anesthesiology 90:1255-1264, 1999

88. Tai YT, Wu CC, Wu GJ, et al: Study of propofol in bovine aortic endothelium: I. Inhibitory effect on bradykinin-induced intracellular calcium immobilization. Acta Anesthesiol Sinica 38:181-186, 2000

(8)

89. Park WK, Lynch C 3rd, Johns RA: Effects of propofol and thiopental in isolated rat aorta and pulmonary artery. Anesthesiology 77:956-963, 1992

90. Gacar N, Gök S, Kalyoncu NI, et al: The effect of endothelium on the response to propofol on bovine coronary artery rings. Acta Anaesthesiol Scand 39:1080-1083, 1995

91. Coughlan MG, Flynn NM, Kenny D, et al: Differential relaxant effect of high concentrations of intravenous anesthetics on endothelin-constricted proximal and distal canine coronary arteries. Anesth Analg 74:378-383, 1992

92. Klockgether-Radke AP, Frerichs A, Kettler D, et al: Propofol and thiopental attenuate the contractile response to vasoconstrictors in human and porcine coronary artery segments. Eur J Aneshesiol 17:485-490, 2000

93. Klockgether-Radke AP, Schulze H, Neumann P, et al: Activa-tion of the K⫹ channel BK(Ca) is involved in the relaxing effect of propofol on coronary arteries. Eur J Anaesthesiol 21:226-230, 2004

94. Wallerstedt SM, Törnebrandt K, Bodelsson M: Relaxant effects of propofol on human omental arteries and veins. Br J Anaesth 80:655-659, 1998

95. Bodelsson G, Sandstrom K, Wallerstedt SM, et al: Effects of propofol on substance P-induced relaxation in isolated human omental arteries and veins. Eur J Anesthesiol 17:720-728, 2000

96. Moreno L, Martínez-Cuesta MA, Muedra V, et al: Role of the endothelium in the relaxation induced by propofol and thiopental in isolated arteries from man. J Pharm Pharmacol 49:430-432, 1997

97. Gursoy S, Berkan O, Bagcivan I, et al: Effects of intravenous anesthetics on the human radial artery used as a coronary artery bypass graft. J Cardiothorac Vasc Anesth 21:41-44, 2007

98. Park KW, Dai HB, Lowenstein E, et al: Propofol-associated dilation of rat distal coronary arteries is mediated by multiple sub-stances, including endothelium-derived nitric oxide. Anesth Analg 81: 1191-1196, 1995

99. Horibe M, Kondo I, Damron DS, et al: Propofol attenuates capacitative calcium entry in pulmonary artery smooth muscle cells. Anesthesiology 95:681-688, 2001

100. Boillot A, Laurant P, Berhelot A, et al: Effects of propofol on vascular reactivity in isolated aortae from normotensive and spontane-ously hypertensive rats. Br J Anaesth 83:622-629, 1999

101. Ogawa K, Tanaka S, Murray PA: Propofol potentiates phen-ylephrine-induced contraction via cyclooxygenase inhibition in pulmo-nary artery smooth muscle. Anesthesiology 94:833-839, 2001

102. Kondo U, Kim SO, Murray PA: Propofol selectively attenuates endothelium-dependent pulmonary vasodilation in chronically instru-mented dogs. Anesthesiology 93:437-446, 2000

103. Tanaka H, Yamanoue T, Kroba M, et al: Propofol relaxes extrapulmonary artery but not intrapulmonary artery through nitric oxide pathway. Hiroshima J Med Sci 50:61-64, 2001

104. Horibe M, Ogawa K, Sohn JT, et al: Propofol attenuates acetylcholine-induced pulmonary vasorelaxation: Role of nitric oxide and endothelium-derived hyperpolarizing factors. Anesthesiology 93: 447-455, 2000

Riferimenti

Documenti correlati

La motivazione che mi ha spinto a sviluppare questo tema è avvenuta durante il mio stage in un servizio ambulatoriale di psichiatria. Osservai che tra gli utenti era un tema

Other potential mechanisms that have been put forward to explain the insulin resistance, include increased lipid oxidation and glucose toxicity (DeFronzo, 1992)

Complessivamente, dunque, questo lavoro si propone di ricostruire a grandi linee quel che accade attorno al “muro della vergogna”, a partire dalla constatazione che la

L’adozione di sistemi di calcolo del peso assi- stenziale dei pazienti permette di potenziare per esempio, le presenze in turno, quando vi sono uno o più pazienti in

Dall'autore di 'La maledizione dei Lawrence' Episodio 8 (conclusivo) Un giovane ispettore, catapultato in un paesino di sole sette. Compra Ebook Un ritorno inaspettato -

utilizzo degli indicatori di efficienza e di qualità del Servizio Sanitario Nazionale”. 14) CDC/NHSN “Surveillance definitions for specific Types of Infections” January 2014;1-

Arnow nasce in Kentucky nel 1907 e, nonostante trascorra gran parte della sua vita in diverse città del Nord, in particolare a Detroit, ha ambientato quasi tutti i suoi romanzi