Skip to main content
F1000Research logoLink to F1000Research
. 2015 Aug 25;4(F1000 Faculty Rev):626. [Version 1] doi: 10.12688/f1000research.6288.1

Sevoflurane

Stefan De Hert 1,a, Anneliese Moerman 1
PMCID: PMC4560253  PMID: 26380072

Abstract

Sevoflurane has been available for clinical practice for about 20 years. Nowadays, its pharmacodynamic and pharmacokinetic properties together with its absence of major adverse side effects on the different organ systems have made this drug accepted worldwide as a safe and reliable anesthetic agent for clinical practice in various settings.

Keywords: Sevoflurane, anesthetic, anesthesiology, pharmacodynamic, pharmacokinetic

Introduction

Although sevoflurane was synthesized in the early 1970s 1, it was not released for clinical use until the early 1990s. This was related partly to the expensive synthesis and the initial concern of apparent toxic effects 2, which later appeared to be a consequence of a flawed experimental design 3. Nowadays, its pharmacodynamic and pharmacokinetic properties together with its absence of major adverse side effects on the different organ systems have made this drug accepted worldwide as a safe and reliable anesthetic agent for clinical practice in various settings.

Physicochemical properties

Sevoflurane (1,1,1,3,3,3-hexafluoro-2-(fluoromethoxy)propane) is a colorless, volatile, and non-flammable liquid with a characteristic smell. It is stable at room temperature and has a boiling point of 58.6°C and a vapor pressure of 157 mm Hg. Hence, in contrast to desflurane, it can be used in standard vaporizers 3. Sevoflurane has an oil/gas partition coefficient of 47.2 and its minimal alveolar concentration (MAC), which is the percentage that is necessary to prevent movement in 50% of patients during skin incision, is 2.05% 4, 5. As a consequence, its potency is considerably lower than that of the older inhalational agents such as halothane and isoflurane, but it is about three times more potent than desflurane.

Upon contact with alkaline carbon dioxide (CO 2) absorbers, sevoflurane undergoes degradation 68. The most important degradation product is fluoromethyl-2,2-difluoro-1-(trifluoromethyl) vinyl ether, better known as compound A. In experimental studies, compound A has been reported to be nephrotoxic 9, 10. Although the clinical implications of these findings remained unclear 11, the safety issue related to compound A formation led to intense debates for many years before the issue was resolved 12.

In 1996, Abbott Laboratories voluntarily recalled one lot of sevoflurane because evaluation of a bottle of sevoflurane revealed an uncharacteristically pungent odor 13, 14. This was caused by the formation of hydrogen fluoride as a consequence of a Lewis acid-fluorocarbon reaction. Even in minute amounts, this substance is highly reactive and toxic and can cause respiratory irritation and pulmonary hemorrhage 15. Subsequently, the offending Lewis acid (ferric oxide)-containing part was removed from the sevoflurane handling equipment, and a Lewis acid inhibitor (water) was added to the final product 16, 17. Although Abbott adapted the production process to create a “high water” sevoflurane (>300 parts per million), the manufacturers that subsequently launched sevoflurane (Minrad and Baxter) did not 1820.

Pharmacological properties

It is beyond the scope of this review to discuss in detail the pharmacological properties of sevoflurane. Several excellent review articles have addressed this topic 2125.

Pharmacodynamics

MAC values of sevoflurane decrease with age, from 3.3% in neonates and 2.5% in infants and young adults to 1.58% to 2.05% in middle-aged adults and 1.45% in adults who are more than 70 years old 2631. In the presence of 65% nitrous oxide in the inspired gas mixture, MAC values for sevoflurane decrease by about 50% in adults 32.

Gender does not influence the MAC of sevoflurane, but there is some evidence suggesting that ethnic factors may play a role: MAC values reported in US studies were consistently higher (2.05% to 2.6%) 5, 32 than those reported for Japanese adults (1.58% to 1.71%) 30, 31.

Pharmacokinetics

As for the volatile anesthetic uptake, distribution and elimination are best described by a five-compartment mammillary model 33. This model consists of the lungs, the vessel-rich group of organs, muscle, fat adjacent to the vessel-rich organs, and peripheral fat. In general, an inverse relationship exists between the blood/gas partition coefficient of a volatile anesthetic and the time required for the inspired and alveolar concentrations to reach equilibrium. Sevoflurane has a low blood/gas partition coefficient (0.69), resulting in a swifter equilibration of the alveolar-to-inspiratory fraction (F A/F I ratio) than with enflurane and isoflurane but slightly slower than with nitrous oxide and desflurane 33, 34.

Because of its pleasant odor and the absence of irritation to the airways, sevoflurane can be used for inhalational induction both in children and in adults 35. Studies have shown that induction is as rapid as 36, 37 or even swifter than 3840 with halothane.

Elimination of a volatile anesthetic is also related to its blood solubility. Between 95% and 98% of sevoflurane is eliminated through the lung. The driving force for this elimination is the difference in partial pressures between the inspired gas mix and the pulmonary capillary blood. In humans, 2% to 5% of the absorbed dose of sevoflurane is metabolized by the liver, resulting in the formation of inorganic fluoride and the organic fluoride metabolite hexafluoroisopropanol 41. The latter is conjugated with glucuronic acid and excreted rapidly via the kidneys. The biotransformation of sevoflurane occurs predominantly through cytochrome P450(CYP)2E1 42, 43. Serum inorganic fluoride concentrations after sevoflurane anesthesia are dose-dependent, reaching 10 to 20 µmol/L after 1 to 2 MAC hours and up to 20 to 90 µmol/L with prolonged exposure 41. Although most studies could not show nephrotoxic effects after sevoflurane anesthesia 44, some controversial reports 45 of mild renal dysfunction after the use of sevoflurane resulted in a recommendation by the US Food and Drug Administration for caution in the use of sevoflurane in patients with coexisting renal disease. Interestingly, the majority of data report no differences in pharmacokinetics between patients with and those without kidney diseases 46, 47. Percutaneous losses account for less than 1% of the total uptake of sevoflurane 48.

Effects on vital systems

Like the effects of other anesthetic agents, those of sevoflurane on the vital systems are mostly depressant.

Respiration

A decrease in ventilation leading to apnea at concentrations of between 1.5 and 2.0 MAC can be observed. The ventilatory depression with sevoflurane is the result of a combination of central depression of medullary respiratory neurons 49 and depression of diaphragmatic function 50 and contractility 51.

Sevoflurane provides bronchodilation and attenuates bronchial smooth muscle constriction by histamine or acetylcholine and can be safely used in patients with asthma 21. Hypoxic pulmonary vasoconstriction is inhibited by sevoflurane in a dose-dependent manner and is not mediated by cyclo-oxygenase 2123.

Circulation

Sevoflurane decreases blood pressure in a dose-dependent manner by decreasing total peripheral resistance. At clinically relevant concentrations, cardiac output is usually preserved 2123. Heart rate remains unchanged or even decreases. Coronary blood flow remains preserved and regional blood flow to other vascular beds appears to be maintained at least when systemic hemodynamics are preserved. For sevoflurane (unlike for desflurane), no sympathetic nervous system activation is observed 2123. Although sevoflurane has been reported to prolong the QT and the QTc interval 52, it has no effect on the normal cardiac conduction pathways and therefore is considered a safe agent that can also be used in cardiac electrophysiological procedures 25.

Central nervous system

Sevoflurane is a cerebral vasodilator. In neurosurgical patients, sevoflurane decreased middle cerebral artery flow velocity and caused no epileptiform electroencephalogram activity and no increase of intracranial pressure 53. Cerebral autoregulation is maintained at low concentrations of sevoflurane 54, but higher doses seem to decrease autoregulatory capacity 55.

Safety

Overall, sevoflurane is considered to be a safe and reliable agent that has also been used in uncommon medical conditions such as pheochromocytoma, acute intermittent porphyria, carnitine deficiency, muscular dystrophy, multiple sclerosis, primary aldosteronism, and myotonic dystrophy 23.

In the early years of its use, a number of reports on malignant hyperthermia with sevoflurane were published. In many of them, it was difficult to isolate the potential effects of sevoflurane from the influence of the concurrent use of other triggers such as succinylcholine. Animal studies have suggested that the malignant hyperthermia trigger of sevoflurane was substantially lower than that of other volatile anesthetic agents 56. However, a recent Japanese database study did not find evidence that sevoflurane would be a weaker triggering agent for malignant hyperthermia 57. Since its introduction in clinical practice, sevoflurane has been safely used in millions of people, and reports of sevoflurane-related malignant hyperthermia are scarce. Nevertheless, it seems wise to avoid exposure to sevoflurane in patients with a known susceptibility.

In the early years of clinical sevoflurane use, it was reported that sevoflurane in the presence of the CO 2 absorbers soda lime (calcium, sodium, and potassium hydroxide mixture), or baralyme (barium, sodium, calcium, and potassium hydroxide mixture) degrades to compound A. This degradation occurs in the anesthesia machine as a result of the extraction of an acidic proton (from the inhalational anesthetic) by a strong base (soda lime or baralyme) 58. The rate of degradation at a given temperature and moisture level is two to four times greater with baralyme compared with soda lime 58, 59. The order of solubility of inhalation anesthetics in dry soda lime is sevoflurane > enflurane > desflurane ≥ halothane > isoflurane 4. As a consequence, more sevoflurane is absorbed into the CO 2 absorber than is observed with other inhalational anesthetics. The production and subsequent inhalation of compound A correlate inversely with the inflow rate 60 and directly with the absorbent temperature 61. In addition, low fresh gas flows of sevoflurane are associated with increased temperatures in the CO 2 absorber 62. Therefore, compound A production can be limited by decreasing the temperature of the absorbent 63. For these reasons, US and Canadian package labels and licensing authorities have recommended minimal fresh gas inflow rates of 1 or 2 L/min, although other licensing authorities have not made such a recommendation. Compound A production can also be reduced by the amount of absorbent (smaller canisters) 64 and adapting the composition of the absorbent by eliminating potassium and sodium hydroxide 6567. The clinical implications of compound A production have been a point of debate for many years 68, but the introduction of the new-generation absorbers 11 has made this issue largely obsolete.

Special populations

The pediatric patient

There seems to be no significant difference in sevoflurane pharmacokinetics between children and adults 24. Because of its pleasant odor, lack of airway irritation, and maintenance of stable hemodynamics, sevoflurane is the agent of choice for mask induction. In general, complications upon emergence are infrequent, although some studies mention a significantly higher incidence of excitement/agitation with sevoflurane 69, 70. However, this observation has been linked to the fact that the prompt recovery from anesthesia with sevoflurane also facilitates earlier awareness of postoperative pain 69, 71. This causal relationship was confirmed in a number of studies demonstrating that adequate pain treatment was associated with significantly fewer episodes of emergence agitation 72, 73.

The ambulatory patient

Ambulatory surgery has increased rapidly in recent years and this has put an emphasis on the use of short-acting drugs in anesthetic practice, allowing fast recovery and early mobilization. Differences in early recovery between sevoflurane, desflurane, and propofol have been reported to be small but in favor of the inhaled anesthetics 74, although the clinical implications of these small differences are debatable. Postoperative nausea and vomiting are higher with volatile anesthetics than with propofol, but adequate anti-emetic prophylaxis can prevent or blunt this side effect.

The obese patient

The prevalence of obesity is increasing dramatically, not only in industrialized countries but also in developing ones. As a consequence, we encounter a growing number of morbidly obese patients who need different types of surgery. Obese patients are traditionally reported to have slower emergence from anesthesia because of a delayed release of volatile anesthetics from the excess fat tissue. However, comparable recovery times have been reported in obese and non-obese subjects after anesthetic procedures lasting 2 to 4 hours 75.

The new inhalation drugs have a much lower lipid solubility compared with the older volatile anesthetic agents, resulting in a more rapid and consistent recovery profile 76. For sevoflurane, no significant differences in F A/F I ratio have been observed, but the wash-out curve—that is, the alveolar-to-expiratory fraction (F A/F AO ratio)—was reported to be slower in obese patients compared with non-obese patients. However, 5 minutes after sevoflurane discontinuation, no differences in wash-out were observed between obese and non-obese patients 77. Several studies have compared kinetic profiles of sevoflurane and desflurane. Some studies observed more rapid emergence from anesthesia with desflurane but this was not confirmed in other studies (reviewed in 78). Finally, an advantage of sevoflurane in this setting is that it allows progressive induction of anesthesia via face mask 79.

Organ protection

The heart

The finding in the late 1990s that sevoflurane was capable of limiting the extent of myocardial infarction after myocardial ischemia triggered a new research direction investigating potential cardioprotective effects of volatile anesthetic agents. Experimental studies have clearly indicated that volatile anesthetic agents are capable of protecting the myocardium against the consequences of ischemia-reperfusion injury by decreasing the extent of myocardial damage, decreasing the extent of reperfusion injury, and better preserving myocardial function. Subsequent research was directed toward unraveling the underlying mechanisms and intracellular pathways of these cardioprotective effects 8085.

Although the experimental evidence of cardioprotection with volatile anesthetic agents was quite straightforward, the implications for clinical practice remain a point of debate. The potential cardioprotective effects related to the use of volatile anesthetics were first explored in the setting of cardiac surgery. In coronary artery surgery patients, the results of preconditioning protocols were conflicting: some authors demonstrated a protective effect whereas others failed to observe such an effect. Later, it became clear that this might be attributed to the preconditioning protocol used. It also seems that the administration of the volatile anesthetic agent throughout the entire procedure results in a more pronounced protective effect than when administered intermittently. It is beyond the scope of this review to discuss these studies in detail. The interested reader is referred to a number of reviews on the topic 8695.

For non-cardiac surgery, the potential clinical implication of the cardioprotective properties of volatile anesthetics is even more debatable. One small study in vascular surgery patients observed a lower incidence of cardiac complications in patients treated with sevoflurane compared with those anesthetized with propofol 96. Others, however, observed no difference in the extent of myocardial damage when comparing a volatile anesthetic regimen with a total intravenous regimen 97, 98. This clearly indicates that only in the presence of myocardial ischemia/reperfusion injury can a potential beneficial effect of volatile anesthetics be expected 99. Interestingly, in the study by Lurati Buse and colleagues 98, in which 385 patients were randomly assigned to receive anesthesia with either sevoflurane or propofol, the incidences of perioperative myocardial ischemia were comparable (40.8% in the sevoflurane group and 40.3% in the propofol group). Within 12 months, 14 patients had a major cardiac event in the sevoflurane-treated group (7.6%) and 17 in the propofol-treated group (8.5%). However, given that a potential cardioprotective effect of volatile anesthetic agents relates to a modulation of the extent of myocardial ischemia/reperfusion injury, the analysis of the major cardiac events needs to be focused on the occurrence of these events in the subgroup of patients who had perioperative myocardial ischemia. To further clarify this issue, the authors performed an additional analysis of their data. They found that the incidence of major cardiac complications in patients with evidence of perioperative myocardial ischemia was similar in both groups: 8 out of 67 patients (11.9%) in the sevoflurane group and 9 out of 72 patients (12.9%) in the propofol group. The remaining 6 and 8 patients with postoperative major cardiac complications had not shown any evidence of perioperative myocardial ischemia (Seeberger M, unpublished observations).

Other organ systems

Clinical studies on the protective effects of sevoflurane on other organ systems are scarce and limited to a small number of patients. Three studies from the same group suggest protective effects after liver 100, 101 and lung 102 ischemia with sevoflurane, but the potential implications on long-term outcome remain to be established.

Neurotoxicity in the young and aged brain

Preclinical evidence in rodents and non-human primates has caused concern regarding the safety of anesthesia in infants and children. Indeed, animal studies suggest that neurodegeneration with possible cognitive sequelae may constitute a potential long-term risk of anesthesia in neonatal and young pediatric patients (reviewed in 103, 104). No hard clinical data suggest that the use of anesthetics in the neonate or young child is associated with signs of developmental neurotoxicity 103. It has been argued that the increased risk of poor outcome in some human cohort studies is because of the inflammation and stress associated with the surgery rather than the anesthetic 105. It is expected that the results of two ongoing large-scale studies—the Multi-site Randomized Controlled Trial comparing Regional and General Anesthesia for Effects on Neurodevelopment Outcome and Apnea in Infants (GAS) study and the Pediatric Anesthesia and NeuroDevelopment Assessment (PANDA) study—will give more insight into the problem 104.

Similarly, experimental studies, observing effects of anesthetic agents on memory formation and the induction of neurodegenerative changes on a cellular level, have raised concerns about the effects of anesthesia and surgery on the elderly brain. The incidence of postoperative cognitive dysfunction (POCD) varies according to the definitions used in the various studies but is reported to be higher in major surgery (reviewed in 106108). Whether general anesthesia contributes to POCD remains uncertain. A recent meta-analysis of 26 randomized trials comparing general to regional anesthesia was unable to identify general anesthesia as an independent risk factor for POCD 109. It is conceivable that surgical trauma and underlying pathology are of greater importance. Given the complexity and still-unknown elements of the pathogenesis of POCD, further research on the topic is needed.

Conclusions

Since its introduction in clinical practice, sevoflurane has gained wide acceptance as an anesthetic for various types of surgery. Its ease of administration, versatility, and stable hemodynamic profile make it a safe and easily applicable anesthetic agent.

Funding Statement

The author(s) declared that no grants were involved in supporting this work.

v1; ref status: indexed

References

  • 1. Wallin RF, Regan BM, Napoli MD, et al. : Sevoflurane: a new inhalational anesthetic agent. Anesth Analg. 1975;54(6):758–66. 10.1213/00000539-197511000-00021 [DOI] [PubMed] [Google Scholar]
  • 2. Hitt B, Mazze R, Cook T, et al. : Thermoregulatory defect in rats during anesthesia. Anesth Analg. 1977;56(1):9–15. 10.1213/00000539-197701000-00006 [DOI] [PubMed] [Google Scholar]
  • 3. Patel SS, Goa KL: Sevoflurane. A review of its pharmacodynamic and pharmacokinetic properties and its clinical use in general anaesthesia. Drugs. 1996;51(4):658–700. 10.2165/00003495-199651040-00009 [DOI] [PubMed] [Google Scholar]
  • 4. Strum DP, Eger EI 2nd: Partition coefficients for sevoflurane in human blood, saline, and olive oil. Anesth Analg. 1987;66(7):654–6. 10.1213/00000539-198707000-00013 [DOI] [PubMed] [Google Scholar]
  • 5. Scheller MS, Saidman LJ, Partridge BL: MAC of sevoflurane in humans and the New Zealand white rabbit. Can J Anaesth. 1988;35(2):153–6. 10.1007/BF03010656 [DOI] [PubMed] [Google Scholar]
  • 6. Munday IT, Ward PM, Foden ND, et al. : Sevoflurane degradation by soda lime in a circle breathing system. Anaesthesia. 1996;51(7):622–6. 10.1111/j.1365-2044.1996.tb07842.x [DOI] [PubMed] [Google Scholar]
  • 7. Cunningham DD, Huang S, Webster J, et al. : Sevoflurane degradation to compound A in anaesthesia breathing systems. Br J Anaesth. 1996;77(4):537–43. 10.1093/bja/77.4.537 [DOI] [PubMed] [Google Scholar]
  • 8. Eger EI 2nd, Ionescu P, Laster MJ, et al. : Baralyme dehydration increases and soda lime dehydration decreases the concentration of compound A resulting from sevoflurane degradation in a standard anesthetic circuit. Anesth Analg. 1997;85(4):892–8. 10.1097/00000539-199710000-00033 [DOI] [PubMed] [Google Scholar]
  • 9. Gonsowski CT, Laster MJ, Eger EI 2nd, et al. : Toxicity of compound A in rats. Effect of a 3-hour administration. Anesthesiology. 1994;80(3):556–65. 10.1097/00000542-199403000-00012 [DOI] [PubMed] [Google Scholar]
  • 10. Gonsowski CT, Laster MJ, Eger EI 2nd, et al. : Toxicity of compound A in rats. Effect of increasing duration of administration. Anesthesiology. 1994;80(3):566–73. 10.1097/00000542-199403000-00013 [DOI] [PubMed] [Google Scholar]
  • 11. Marini F, Bellugi I, Gambi D, et al. : Compound A, formaldehyde and methanol concentrations during low-flow sevoflurane anaesthesia: comparison of three carbon dioxide absorbers. Acta Anaesthesiol Scand. 2007;51(5):625–32. 10.1111/j.1399-6576.2007.01278.x [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 12. Gentz BA, Malan TP, Jr: Renal toxicity with sevoflurane: a storm in a teacup? Drugs. 2001;61(15):2155–62. 10.2165/00003495-200161150-00001 [DOI] [PubMed] [Google Scholar]
  • 13. Leary JP: Contaminated sevoflurane use reported from NY state. APSF Newsletter. 1996;11:37 Reference Source [Google Scholar]
  • 14. Callan CM: Sevo manufacturer outlines circumstances, response. APSF Newsletter. 1996;11:37 Reference Source [Google Scholar]
  • 15. Bertolini J: Hydrofluoric acid: a review of toxicity. J Emerg Med. 1992;10(2):163–8. 10.1016/0736-4679(92)90211-B [DOI] [PubMed] [Google Scholar]
  • 16. McLeskey CH: Anesthesiologist executive reports how Abbott made sevoflurane safer: water stops formation of highly toxic acid. APSF Newsletter. 2000;15:39 Reference Source [Google Scholar]
  • 17. Kharash ED, Subbarao GN, Cromack KR, et al. : Sevoflurane formulation water content influences degradation by Lewis acids in vaporizers. Anesth Analg. 2009;108(6):1796–802. 10.1213/ane.0b013e3181a3d72b [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 18. Kharash ED: Sevoflurane: the challenges of safe formulation. APSF Newsletter. 2007;22:42 Reference Source [Google Scholar]
  • 19. Baker MT: Sevoflurane: are there differences in products? Anesth Analg. 2007;104(6):1447–51. 10.1213/01.ane.0000263031.96011.36 [DOI] [PubMed] [Google Scholar]
  • 20. Baker MT: Sevoflurane-Lewis acid stability. Anesth Analg. 2009;108(6):1725–6. 10.1213/ane.0b013e3181a72d07 [DOI] [PubMed] [Google Scholar]
  • 21. Eger EI 2nd: New inhaled anesthetics. Anesthesiology. 1994;80(4):906–22. 10.1097/00000542-199404000-00024 [DOI] [PubMed] [Google Scholar]
  • 22. Frink EJ, Brown BR: Sevoflurane. Baillieres Clin Anaesthesiol. 1993;7(4):899–913. 10.1016/S0950-3501(05)80154-0 [DOI] [Google Scholar]
  • 23. Young CJ, Apfelbaum JL: Inhalational anesthetics: desflurane and sevoflurane. J Clin Anesth. 1995;7(7):564–77. 10.1016/0952-8180(95)00129-8 [DOI] [PubMed] [Google Scholar]
  • 24. Behne M, Wilke HJ, Harder S: Clinical pharmacokinetics of sevoflurane. Clin Pharmacokinet. 1999;36(1):13–26. 10.2165/00003088-199936010-00002 [DOI] [PubMed] [Google Scholar]
  • 25. Delgado-Herrera L, Ostroff RD, Rogers SA: Sevoflurane: approaching the ideal inhalational anesthetic. A pharmacologic, pharmacoeconomic, and clinical review. CNS Drug Rev. 2001;7(1):48–120. 10.1111/j.1527-3458.2001.tb00190.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Mapleson WW: Effect of age on MAC in humans: a meta-analysis. Br J Anaesth. 1996;76(2):179–85. 10.1093/bja/76.2.179 [DOI] [PubMed] [Google Scholar]
  • 27. Katoh T, Ikeda K: Minimum alveolar concentration of sevoflurane in children. Br J Anaesth. 1992;68(2):139–41. 10.1093/bja/68.2.139 [DOI] [PubMed] [Google Scholar]
  • 28. Inomata S, Watanabe S, Taguchi M, et al. : End-tidal sevoflurane concentration for tracheal intubation and minimum alveolar concentration in pediatric patients. Anesthesiology. 1994;80(1):93–6. 10.1097/00000542-199401000-00016 [DOI] [PubMed] [Google Scholar]
  • 29. Lerman J, Sikich N, Kleinman S, et al. : The pharmacology of sevoflurane in infants and children. Anesthesiology. 1994;80(4):814–24. [DOI] [PubMed] [Google Scholar]
  • 30. Katoh T, Ikeda K: The minimum alveolar concentration (MAC) of sevoflurane in humans. Anesthesiology. 1987;66(3):301–3. 10.1097/00000542-198703000-00006 [DOI] [PubMed] [Google Scholar]
  • 31. Kimura T, Watanabe S, Asakura N, et al. : Determination of end-tidal sevoflurane concentration for tracheal intubation and minimum alveolar anesthetic concentration in adults. Anesth Analg. 1994;79(2):378–81. 10.1213/00000539-199408000-00032 [DOI] [PubMed] [Google Scholar]
  • 32. Fragen RJ, Dunn KL: The minimum alveolar concentration (MAC) of sevoflurane with and without nitrous oxide in elderly versus young adults. J Clin Anesth. 1996;8(5):352–6. 10.1016/0952-8180(96)00082-7 [DOI] [PubMed] [Google Scholar]
  • 33. Yasuda N, Lockhart SH, Eger EI 2nd, et al. : Comparison of kinetics of sevoflurane and isoflurane in humans. Anesth Analg. 1991;72(3):316–24. [DOI] [PubMed] [Google Scholar]
  • 34. Shiraishi Y, Ikeda K: Uptake and biotransformation of sevoflurane in humans: a comparative study of sevoflurane with halothane, enflurane, and isoflurane. J Clin Anaesth. 1990;2(6):381–6. 10.1016/0952-8180(90)90024-W [DOI] [PubMed] [Google Scholar]
  • 35. Doi M, Ikeda K: Airway irritation produced by volatile anaesthetics during brief inhalation: comparison of halothane, enflurane, isoflurane and sevoflurane. Can J Anaesth. 1993;40(2):122–6. 10.1007/BF03011308 [DOI] [PubMed] [Google Scholar]
  • 36. Baum VC, Yemen TA, Baum ID: Immediate 8% sevoflurane induction in children: a comparison with incremental sevoflurane and incremental halothane. Anesth Analg. 1997;85(2):313–6. 10.1213/00000539-199708000-00013 [DOI] [PubMed] [Google Scholar]
  • 37. Sigston PE, Jenkins AM, Jackson EA, et al. : Rapid inhalation induction in children: 8% sevoflurane compared with 5% halothane. Br J Anaesth. 1997;78(4):362–5. 10.1093/bja/78.4.362 [DOI] [PubMed] [Google Scholar]
  • 38. Yurino M, Kimura H: Vital capacity rapid inhalation induction technique: comparison of sevoflurane and halothane. Can J Anaesth. 1993;40(5 Pt 1):440–3. 10.1007/BF03009514 [DOI] [PubMed] [Google Scholar]
  • 39. Yurino M, Kimura H: A comparison of vital capacity breath and tidal breathing techniques for induction of anaesthesia with high sevoflurane concentrations in nitrous oxide and oxygen. Anaesthesia. 1995;50(4):308–11. 10.1111/j.1365-2044.1995.tb04605.x [DOI] [PubMed] [Google Scholar]
  • 40. Nishiyama T, Nagase M, Tamai H, et al. : Rapid induction with 7% sevoflurane inhalation-not the single-breath method. J Anaesth. 1995;9(1):36–9. 10.1007/BF02482033 [DOI] [PubMed] [Google Scholar]
  • 41. Kharash ED: Biotransformation of sevoflurane. Anesth Analg. 1995;81(6 Suppl):S27–38. 10.1097/00000539-199512001-00005 [DOI] [PubMed] [Google Scholar]
  • 42. Kharash ED, Karol MD, Lanni C, et al. : Clinical sevoflurane metabolism and disposition. I. Sevoflurane and metabolite pharmacokinetics. Anesthesiology. 1995;82(6):1369–78. 10.1097/00000542-199506000-00008 [DOI] [PubMed] [Google Scholar]
  • 43. Kharash ED, Armstrong AS, Gunn K, et al. : Clinical sevoflurane metabolism and disposition. II. The role of cytochrome P450 2E1 in fluoride and hexafluoroisopropanol formation. Anesthesiology. 1995;82(6):1379–88. 10.1097/00000542-199506000-00009 [DOI] [PubMed] [Google Scholar]
  • 44. Malan TP, Jr: Sevoflurane and renal function. Anesth Analg. 1995;81(6 Suppl):S39–45. 10.1097/00000539-199512001-00006 [DOI] [PubMed] [Google Scholar]
  • 45. Kharash ED, Hankins DC, Thummel KE: Human kidney methoxyflurane and sevoflurane metabolism. Intrarenal fluoride production as a possible mechanism of methoxyflurane nephrotoxicity. Anesthesiology. 1995;82(3):688–99. 10.1097/00000542-199503000-00011 [DOI] [PubMed] [Google Scholar]
  • 46. Conzen PF, Nuscheler M, Melotte A, et al. : Renal function and serum fluoride concentrations in patients with stable renal insufficiency after anesthesia with sevoflurane or enflurane. Anesth Analg. 1995;81(3):569–75. [DOI] [PubMed] [Google Scholar]
  • 47. Nishiyama T, Aibiki M, Hanaoka K: Inorganic fluoride kinetics and renal tubular function after sevoflurane anesthesia in chronic renal failure patients receiving hemodialysis. Anesth Analg. 1996;83(3):574–7. 10.1097/00000539-199609000-00024 [DOI] [PubMed] [Google Scholar]
  • 48. Lockhart SH, Yasuda N, Peterson N, et al. : Comparison of percutaneous losses of sevoflurane and isoflurane in humans. Anesth Analg. 1991;72(2):212–5. 10.1213/00000539-199102000-00013 [DOI] [PubMed] [Google Scholar]
  • 49. Doi K, Kasaba T, Kosaka Y: [A comparative study of the depressive effects of halothane and sevoflurane on medullary respiratory neurons in cats]. Masui. 1988;37(12):1466–77. [PubMed] [Google Scholar]
  • 50. Ide T, Kochi T, Isono S, et al. : Diaphragmatic function during sevoflurane anaesthesia in dogs. Can J Anaesth. 1991;38(1):116–20. 10.1007/BF03009174 [DOI] [PubMed] [Google Scholar]
  • 51. Ide T, Kochi T, Isono S, et al. : Effect of sevoflurane on diaphragmatic contractility in dogs. Anesth Analg. 1992;74(5):739–46. [DOI] [PubMed] [Google Scholar]
  • 52. Kleinsasser A, Kuenszberg E, Loeckinger A, et al. : Sevoflurane, but not propofol, significantly prolongs the Q-T interval. Anesth Analg. 2000;90(1):25–7. 10.1097/00000539-200001000-00006 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 53. Artru AA, Lam AM, Johnson JO, et al. : Intracranial pressure, middle cerebral artery flow velocity, and plasma inorganic fluoride concentrations in neurosurgical patients receiving sevoflurane or isoflurane. Anesth Analg. 1997;85(3):587–92. 10.1097/00000539-199709000-00019 [DOI] [PubMed] [Google Scholar]
  • 54. Kitaguchi K, Ohsumi H, Kuro M, et al. : Effects of sevoflurane on cerebral circulation and metabolism in patients with ischemic cerebrovascular disease. Anesthesiology. 1993;79(4):704–9. 10.1097/00000542-199310000-00011 [DOI] [PubMed] [Google Scholar]
  • 55. Conti A, Iacopino DG, Fodale V, et al. : Cerebral haemodynamic changes during propofol-remifentanil or sevoflurane anaesthesia: transcranial Doppler study under bispectral index monitoring. Br J Anaesth. 2006;97(3):333–9. 10.1093/bja/ael169 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 56. Kunst G, Graf BM, Schreiner R, et al. : Differential effects of sevoflurane, isoflurane, and halothane on Ca2+ release from the sarcoplasmic reticulum of skeletal muscle. Anesthesiology. 1999;91(1):179–86. 10.1097/00000542-199907000-00026 [DOI] [PubMed] [Google Scholar]
  • 57. Migita T, Mukaida K, Kobayashi M, et al. : The severity of sevoflurane-induced malignant hyperthermia. Acta Anaesthesiol Scand. 2012;56(3):351–6. 10.1111/j.1399-6576.2011.02573.x [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 58. Liu J, Laster MJ, Eger EI 2nd, et al. : Absorption and degradation of sevoflurane and isoflurane in a conventional anesthetic circuit. Anesth Analg. 1991;72(6):785–9. 10.1213/00000539-199106000-00012 [DOI] [PubMed] [Google Scholar]
  • 59. Frink EJ, Jr, Malan TP, Morgan SE, et al. : Quantification of the degradation products of sevoflurane in two CO2 absorbants during low-flow anesthesia in surgical patients. Anesthesiology. 1992;77(6):1064–9. 10.1097/00000542-199212000-00003 [DOI] [PubMed] [Google Scholar]
  • 60. Bito H, Ikeda K: Effect of total flow rate on the concentration of degradation products generated by reaction between sevoflurane and soda lime. Br J Anaesth. 1995;74(6):667–9. 10.1093/bja/74.6.667 [DOI] [PubMed] [Google Scholar]
  • 61. Fang ZX, Kandel L, Laster MJ, et al. : Factors affecting production of compound A from the interaction of sevoflurane with Baralyme® and soda lime. Anesth Analg. 1996;82(4):775–81. 10.1097/00000539-199604000-00018 [DOI] [PubMed] [Google Scholar]
  • 62. Luttropp HH, Johansson A: Soda lime temperatures during low-flow sevoflurane anaesthesia and differences in dead-space. Acta Anaesthesiol Scand. 2002;46(5):500–5. 10.1034/j.1399-6576.2002.460505.x [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 63. Ruzicka JA, Hidalgo JC, Tinker JH, et al. : Inhibition of volatile sevoflurane degradation product formation in an anesthesia circuit by a reduction in soda lime temperature. Anesthesiology. 1994;81(1):238–44. [DOI] [PubMed] [Google Scholar]
  • 64. Yamakage M, Kimura A, Chen X, et al. : Production of compound A under low-flow anesthesia is affected by type of anesthetic machine. Can J Anesth. 2001;48(5):435–8. 10.1007/BF03028304 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 65. Neumann MA, Laster MJ, Weiskopf RB, et al. : The elimination of sodium and potassium hydroxides from desiccated soda lime diminishes degradation of desflurane to carbon monoxide and sevoflurane to compound A but does not compromise carbon dioxide absorption. Anesth Analg. 1999;89(3):768–73. 10.1213/00000539-199909000-00046 [DOI] [PubMed] [Google Scholar]
  • 66. Murray JM, Renfrew CW, Bedi A, et al. : Amsorb: A new carbon dioxide absorbent for use in anesthetic breathing systems. Anesthesiology. 1999;91(5):1342–8. 10.1097/00132586-200010000-00022 [DOI] [PubMed] [Google Scholar]
  • 67. Stabernack CR, Brown R, Laster MJ, et al. : Absorbents differ enormously in their capacity to produce compound A and carbon monoxide. Anesth Analg. 2000;90(6):1428–35. 10.1097/00000539-200006000-00033 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 68. Eger EI 2nd: Compound A: does it matter? Can J Anesth. 2001;48(5):427–30. 10.1007/BF03028302 [DOI] [PubMed] [Google Scholar]
  • 69. Lerman J, Davis PJ, Welborn LG, et al. : Induction, recovery, and safety characteristics of sevoflurane in children undergoing ambulatory surgery.A comparison with halothane. Anesthesiology. 1996;84(6):1332–40. 10.1097/00000542-199606000-00009 [DOI] [PubMed] [Google Scholar]
  • 70. Picard V, Dumont L, Pellegrini M: Quality of recovery in children: sevoflurane versus propofol. Acta Anaesthesiol Scand. 2000;44(3):307–10. 10.1034/j.1399-6576.2000.440315.x [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 71. Welborn LG, Hannallah RS, Norden JM, et al. : Comparison of emergence and recovery characteristics of sevoflurane, desflurane, and halothane in pediatric ambulatory patients. Anesth Analg. 1996;83(5):917–20. 10.1097/00132586-199712000-00004 [DOI] [PubMed] [Google Scholar]
  • 72. Davis PJ, Greenberg JA, Gendelman M, et al. : Recovery characteristics of sevoflurane and halothane in preschool-aged children undergoing bilateral myringotomy and pressure equalization tube insertion. Anesth Analg. 1999;88(1):34–8. 10.1213/00000539-199901000-00007 [DOI] [PubMed] [Google Scholar]
  • 73. Johannesson GP, Florén M, Lindahl SG: Sevoflurane for ENT-surgery in children. A comparison with halothane. Acta Anaesthesiol Scand. 1995;39(4):546–550. 10.1111/j.1399-6576.1995.tb04116.x [DOI] [PubMed] [Google Scholar]
  • 74. Gupta A, Stierer T, Zuckerman R, et al. : Comparison of recovery profile after ambulatory anesthesia with propofol, isoflurane, sevoflurane and desflurane: a systematic review. Anesth Analg. 2004;98(3):632–41. 10.1213/01.ANE.0000103187.70627.57 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 75. Cork RC, Vaughan RW, Bentley JB, et al. : General anesthesia for morbidly obese patients--an examination of postoperative outcomes. Anesthesiology. 1981;54(4):310–3. 10.1097/00132586-198202000-00057 [DOI] [PubMed] [Google Scholar]
  • 76. Sollazzi L, Perilli V, Modesti C, et al. : Volatile anesthesia in bariatric surgery. Obes Surg. 2001;11(5):623–6. 10.1381/09608920160557138 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 77. Casati A, Marchetti C, Spreafico E, et al. : Effects of obesity on wash-in and wash-out kinetics of sevoflurane. Eur J Anaesthesiol. 2004;21(3):243–5. 10.1097/00003643-200403000-00019 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 78. Lemmens HJ: Perioperative pharmacology in morbid obesity. Curr Opin Anaesthesiol. 2010;23(4):485–91. 10.1097/ACO.0b013e32833b0a8c [DOI] [PubMed] [Google Scholar]
  • 79. Salihoglu Z, Karaca S, Kose Y, et al. : Total intravenous anesthesia versus single breath technique and anesthesia maintenance with sevoflurane for bariatric operations. Obes Surg. 2001;11(4):496–501. 10.1381/096089201321209413 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 80. Zaugg M, Lucchinetti E, Uecker M, et al. : Anaesthetics and cardiac preconditioning. Part I. Signalling and cytoprotective mechanisms. Br J Anaesth. 2003;91(4):551–65. 10.1093/bja/aeg205 [DOI] [PubMed] [Google Scholar]
  • 81. Tanaka K, Ludwig LM, Kersten JR, et al. : Mechanisms of cardioprotection by volatile anesthetics. Anesthesiology. 2004;100(3):707–21. 10.1097/00000542-200403000-00035 [DOI] [PubMed] [Google Scholar]
  • 82. Stowe DF, Kevin LG: Cardiac preconditioning by volatile anesthetic agents: a defining role for altered mitochondrial bioenergetics. Antioxid Redox Signal. 2004;6(2):439–48. 10.1089/152308604322899512 [DOI] [PubMed] [Google Scholar]
  • 83. Bienengraeber MW, Weihrauch D, Kersten JR, et al. : Cardioprotection by volatile anesthetics. Vascul Pharmacol. 2005;42(5–6):243–52. 10.1016/j.vph.2005.02.005 [DOI] [PubMed] [Google Scholar]
  • 84. Pratt PF, Jr, Wang C, Weihrauch D, et al. : Cardioprotection by volatile anesthetics: new applications for old drugs? Curr Opin Anaesthesiol. 2006;19(4):397–403. 10.1097/01.aco.0000236139.31099.b5 [DOI] [PubMed] [Google Scholar]
  • 85. Pagel PS, Hudetz JA: Delayed cardioprotection by inhaled anesthetics. J Cardiothorac Vasc Anesth. 2011;25(6):1125–40. 10.1053/j.jvca.2010.09.017 [DOI] [PubMed] [Google Scholar]
  • 86. Zaugg M, Lucchinetti E, Garcia C, et al. : Anaesthetics and cardiac preconditioning. Part II. Clinical implications. Br J Anaesth. 2003;91(4):556–76. 10.1093/bja/aeg206 [DOI] [PubMed] [Google Scholar]
  • 87. De Hert SG, Turani F, Mathur S, et al. : Cardioprotection with volatile anesthetics: mechanisms and clinical implications. Anesth Analg. 2005;100(6):1584–93. 10.1213/01.ANE.0000153483.61170.0C [DOI] [PubMed] [Google Scholar]
  • 88. De Hert SG: The concept of anaesthetic-induced cardioprotection: clinical relevance. Best Pract Res Clin Anaesthesiol. 2005;19(3):445–59. 10.1016/j.bpa.2005.02.004 [DOI] [PubMed] [Google Scholar]
  • 89. De Hert SG: Anesthetic preconditioning: how important is it in today’s cardiac anesthesia? J Cardiothorac Vasc Anesth. 2006;20(4):473–6. 10.1053/j.jvca.2006.05.001 [DOI] [PubMed] [Google Scholar]
  • 90. De Hert S: Cardioprotection in anaesthesia. Minerva Anaesthesiol. 2008;74:259–70. [PubMed] [Google Scholar]
  • 91. De Hert S, Preckel B, Schlack WS: Update on inhalational anaesthetics. Curr Opin Anaesthesiol. 2009;22(4):491–5. 10.1097/ACO.0b013e32832bca38 [DOI] [PubMed] [Google Scholar]
  • 92. Frässdorf J, De Hert S, Schlack W: Anaesthesia and myocardial ischaemia/reperfusion injury. Br J Anaesth. 2009;103(1):89–98. 10.1093/bja/aep141 [DOI] [PubMed] [Google Scholar]
  • 93. De Hert SG: Is anaesthetic cardioprotection clinically relevant? Another futile search for a magic bullet? Eur J Anaesthesiol. 2011;28(9):616–7. 10.1097/EJA.0b013e3283497d00 [DOI] [PubMed] [Google Scholar]
  • 94. Bein B: Clinical application of the cardioprotective effects of volatile anaesthetics: PRO--get an extra benefit from a proven anaesthetic free of charge. Eur J Anaesthesiol. 2011;28(9):620–2. 10.1097/EJA.0b013e328349d5f3 [DOI] [PubMed] [Google Scholar]
  • 95. Van Rompaey N, Barvais L: Clinical application of the cardioprotective effects of volatile anaesthetics: CON--total intravenous anaesthesia or not total intravenous anaesthesia to anaesthetise a cardiac patient? Eur J Anaesthesiol. 2011;28(9):623–7. 10.1097/EJA.0b013e328349aca4 [DOI] [PubMed] [Google Scholar]
  • 96. Van der Linden PJ, Dierick A, Wilmin S, et al. : A randomized controlled trial comparing an intraoperative goal-directed strategy with routine clinical practice in patients undergoing peripheral arterial surgery. Eur J Anaesthesiol. 2010;27(9):788–93. 10.1097/EJA.0b013e32833cb2dd [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 97. Zangrillo A, Testa V, Aldrovandi V, et al. : Volatile agents for cardiac protection in noncardiac surgery: a randomized controlled study. J Cardiothorac Vasc Anesth. 2011;25(6):902–7. 10.1053/j.jvca.2011.06.016 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 98. Lurati Buse GA, Schumacher P, Seeberger E, et al. : Randomized comparison of sevoflurane versus propofol to reduce perioperative myocardial ischemia in patients undergoing noncardiac surgery. Circulation. 2012;126(23):2696–704. 10.1161/CIRCULATIONAHA.112.126144 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 99. De Hert SG: Cardioprotection by volatile anesthetics: what about noncardiac surgery? J Cardiothorac Vasc Anesth. 2011;25(6):899–901. 10.1053/j.jvca.2011.08.004 [DOI] [PubMed] [Google Scholar]
  • 100. Beck-Schimmer B, Breitenstein S, Urech S, et al. : A randomized controlled trial on pharmacological preconditioning in liver surgery using a volatile anesthetic. Ann Surg. 2008;248(6):909–18. [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 101. Beck-Schimmer B, Breitenstein S, Bonvini JM, et al. : Protection of pharmacological postconditioning in liver surgery: results of a prospective randomized controlled trial. Ann Surg. 2012;256(5):837–44; discussion 844–5. 10.1097/SLA.0b013e318272df7c [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 102. De Conno E, Steurer MP, Wittlinger M, et al. : Anesthetic-induced improvement of the inflammatory response to one-lung ventilation. Anesthesiology. 2009;110(6):1316–26. 10.1097/ALN.0b013e3181a10731 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 103. Mellon RD, Simone AF, Rappaport BA: Use of anesthetic agents in neonates and young children. Anesth Analg. 2007;104(3):509–20. 10.1213/01.ane.0000255729.96438.b0 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 104. Sun L: Early childhood general anaesthesia exposure and neurocognitive development. Br J Anaesth. 2010;105(Suppl 1):i61–8. 10.1093/bja/aeq302 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Davidson AJ: Neurotoxicity and the need for anesthesia in the newborn: does the emperor have no clothes? Anesthesiology. 2012;116(3):507–9. 10.1097/ALN.0b013e3182475673 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 106. Hussain M, Berger M, Eckenhoff RG, et al. : General anesthetic and the risk of dementia in elderly patients: current insights. Clin Interv Aging. 2014;9:1619–28. 10.2147/CIA.S49680 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Strøm C, Rasmussen LS, Sieber FE: Should general anaesthesia be avoided in the elderly? Anaesthesia. 2014;69(Suppl 1):35–44. 10.1111/anae.12493 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Mashour GA, Woodrum DT, Avidan MS: Neurological complications of surgery and anaesthesia. Br J Anaesth. 2015;114(2):194–203. 10.1093/bja/aeu296 [DOI] [PubMed] [Google Scholar]
  • 109. Guay J: General anaesthesia does not contribute to long-term post-operative cognitive dysfunction in adults: A meta-analysis. Indian J Anaesth. 2011;55(4):358–63. 10.4103/0019-5049.84850 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
F1000Res. 2015 Aug 25. doi: 10.5256/f1000research.6744.r10111

Referee response for version 1

Manfred Seeberger 1

I have read this submission. I believe that I have an appropriate level of expertise to confirm that it is of an acceptable scientific standard.

F1000Res. 2015 Aug 25. doi: 10.5256/f1000research.6744.r10110

Referee response for version 1

Roderic Eckenhoff 1

I have read this submission. I believe that I have an appropriate level of expertise to confirm that it is of an acceptable scientific standard.


Articles from F1000Research are provided here courtesy of F1000 Research Ltd

RESOURCES