Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Apr 14.
Published in final edited form as: Curr Opin Anaesthesiol. 2022 Jul 5;35(4):425–435. doi: 10.1097/ACO.0000000000001156

Novel anesthetics in pediatric practice: is it time?

Nemanja Useinovic a, Vesna Jevtovic-Todorovic a,b
PMCID: PMC10104442  NIHMSID: NIHMS1884634  PMID: 35787582

Abstract

Purpose of review

Steadily mounting evidence of anesthesia-induced developmental neurotoxicity has been a challenge in pediatric anesthesiology. Considering that presently used anesthetics have, in different animal models, been shown to cause lasting behavioral impairments when administered at the peak of brain development, the nagging question, ‘Is it time for the development of a new anesthetic’ must be pondered.

Recent findings

The emerging ‘soft analogs’ of intravenous anesthetics aim to overcome the shortcomings of currently available clinical drugs. Remimazolam, a novel ester-analog of midazolam, is a well tolerated intravenous drug with beneficial pharmacological properties. Two novel etomidate analogs currently in development are causing less adrenocortical suppression while maintaining equally favorable hemodynamic stability and rapid metabolism. Quaternary lidocaine derivatives are explored as more potent and longer lasting alternatives to currently available local anesthetics. Xenon, a noble gas with anesthetic properties, is being considered as an anesthetic-sparing adjuvant in pediatric population. Finally, alphaxalone is being reevaluated in a new drug formulation because of its favorable pharmacological properties.

Summary

Although a number of exciting anesthetic drugs are under development, there is currently no clear evidence to suggest their lack of neurotoxic properties in young brain. Well designed preclinical studies are needed to evaluate their neurotoxic potential.

Keywords: behavior, children, neuroactive steroids, synaptogenesis, young brain

INTRODUCTION

The requirement for adequate and readily controlled depth of anesthesia during surgical procedures and the foremost importance of patient safety have shifted the anesthetic drug development paradigm. The recent trend towards a ‘soft drug’ design, defined by rapid onset of action and recovery, increased therapeutic index and predictable metabolism to inactive metabolites [1,2], aims to provide the anesthesiologist with a very powerful tool. As the chemical structure of a given drug is intimately linked to its pharmacokinetic properties, this is often achieved via introduction of an ester bond, making the molecule prone to metabolism by nonspecific tissue esterases. On the other hand, the need for long-lasting nerve blocks and effective postoperative pain control call for more potent drugs with longer duration of action with less local and/or systemic toxicity [3].

On the basis of extensive body of preclinical [413] and growing clinical [1419] evidence that early-in-life anesthesia causes widespread and long-lasting neurological damage because of unphysiological activation of neuroapoptosis resulting in permanent reduction in neuronal densities [5] and long-term behavioral impairments [4], novel general and local anesthetics should be designed with anesthesia-induced developmental neurotoxicity in mind. We have recently published a summary of the most promising neuroprotective strategies that may enable well tolerated use of currently available anesthetics [20]. Here, we highlight the recent advances in anesthetic drug development and evaluate their potential for clinical use in children.

REMIMAZOLAM: A NOVEL ULTRA-SHORTACTING BENZODIAZEPINE

The intravenous anesthesia and sedation in the current practice are predominantly achieved with propofol and/or midazolam. The slow onset of action and presence of active metabolite along with certain controversies about the quality and depth of midazolam-induced anesthesia [21,22] have diminished its use in favor of propofol, which provides faster onset of hypnosis, rapid recovery, antiemetic properties and enables target-controlled infusion (TCI). Notwithstanding these advantages, the narrow therapeutic index, systemic hypotension and central apnea, pain on injection site, phlebitis, and lipid accumulation associated with prolonged use of propofol have brought up a consideration for the development of potentially better intravenous anesthetics.

Remimazolam (CNS7056) is a novel ultra-short acting benzodiazepine [23]. The structure of remimazolam is almost identical to midazolam with the addition of an ester bond in the side chain. As a result, remimazolam undergoes metabolism by nonspecific tissue esterases, making it a useful drug in hepatic or renal failure. Remimazolam was approved for use as a general anesthetic in Japan in 2020 and for procedural anesthesia in the USA in 2021.

The ultra-rapid pharmacokinetics of remimazolam [23] have triggered several clinical trials aiming to revisit the concept of benzodiazepine-based general anesthesia. Recently, two large multicenter randomized controlled trials have been conducted to evaluate the feasibility of remimazolam versus propofol-based anesthesia in low-risk [24■■] and high-risk patients [25■■]. Both studies concluded that remimazolam has excellent ability to both induce and maintain anesthesia with fewer hypotensive episodes compared with propofol. Although remimazolam-based anesthesia resulted in slightly longer overall duration of a procedure because of longer latency to loss of consciousness and longer time to extubation [24■■], as well as delayed time to discharge from the operating room compared with propofol, this could be because of technical difficulties rather than the intrinsic properties of the drug. Of note, flumazenil administration to two patients with delayed recovery resulted in the immediate return of consciousness [25■■].

The intrinsic hemodynamic stability of remimazolam could be of particular value in patients with reduced cardiovascular reserve. In patients undergoing valve replacement surgery, induction of with remimazolam resulted in significantly fewer hypotensive episodes and a decreased use of vasopressors [26]. Remimazolam was also suitable for induction in elderly patients with severe aortic stenosis with no serious adverse effects [27], suggesting that remimazolam could be beneficial in patients with compromised cardiovascular function.

In the United States, remimazolam is registered as a sedative for procedural anesthesia. Sedation with remimazolam versus midazolam or placebo led to superior procedural success rates in bronchoscopy and gastrointestinal endoscopy [2830] cases with shorter length of procedure and faster and more complete return to preprocedure cognitive functions. Recently, it has been reported that female patients undergoing hysteroscopy exhibited fewer adverse effects and less respiratory depression with remimazolam compared with propofol sedation with improved overall recovery [31].

Currently, the preclinical literature involving remimazolam administration is scarce. Through TLR4 receptor downregulation, remimazolam was shown to interfere with lipopolysaccharide (LPS)-induced endotoxemia via blockade of NF-kB signaling pathway and reversal of LPS-induced phosphorylation of Akt and Erk protein kinases essential for neuronal survival [32■■]. The interaction with NF-kB was also beneficial in promoting glioma cell apoptosis in vitro [33] and in alleviating neuropathic pain in adult rats [34].

Remimazolam, like midazolam, is a potent GABAA receptor agonist [23], which is the hallmark property of many neurotoxic anesthetics [4,3539]. Interestingly, there is a report suggesting that remimazolam may negatively affect cognition in the elderly [40]. On the basis of the pharmacodynamic properties of remimazolam, there is currently no evidence to suggest it may be any different than other GABAA-agonists with regard to anesthesia-induced neurotoxicity. However, the rapid conversion to completely inactive metabolites [23], easier dose titration and a potential for pharmacological reversal might reduce the cumulative neurotoxicity compared with other clinically available anesthetics. Of note, remimazolam was safely applied in 3-year-old Cynomolgus monkeys [41] and in a 4-year-old child with Duchenne muscular dystrophy undergoing inguinal herniorrhaphy [42]. Therefore, we believe that well designed preclinical studies in young animals aiming to compare the neurotoxic potential of remimazolam versus commonly used anesthetics, such as propofol or midazolam, would be a valuable addition to the current literature.

ETOMIDATE ANALOGS: A QUEST FOR CARDIOVASCULAR AND ADRENAL SAFEGUARD

The original enthusiasm towards etomidate as a rapidly acting anesthetic with wide therapeutic index and excellent cardiovascular stability had significantly diminished when increased mortality following etomidate-based anesthesia was observed [43]. As a potent inhibitor of 11β-hydroxylase [44,45], a rate-limiting step in steroid synthesis, etomidate use can lead to adrenocortical suppression for over a day even after routine application, resulting in almost complete abandonment of its clinical use.

The interest for etomidate was renewed after significant efforts were made to develop analog that would result in less adrenocortical suppression while preserving the rapid kinetics and hemodynamic stability. Cyclopropyl-methoxycarbonylmetomidate (CPMM) has emerged among many candidate drugs as a new ‘soft’ etomidate ester-analog. In contrast with etomidate, anesthesia with CPMM in LPS-induced sepsis model in rats was associated with less adrenocortical suppression, lower proinflammatory cytokine levels and less mortality [46] suggesting that CPMM might be the superior anesthetic for use in critically ill patients. Although relatively few, studies on healthy volunteers suggest that CPMM is well tolerated with few mild side effects [47,48]. Of note, involuntary muscle movements were present in a dose-dependent fashion, but they were not ictal in origin [49] and were effectively treated with midazolam or fentanyl [47,48].

A different line of research developed ET-26 hydrochloride (ET-26HCl), another etomidate analog currently approved for clinical trials in China. Early research showed that ET-26HCl preserved the rapid onset and hemodynamic stability of parent compound etomidate with very little adrenocortical suppression, which was comparable to that of CPMM [50,51]. The hemodynamic and adrenal stability together with superior myocardial performance following ET-26HCl administration [52] were demonstrated in rat models of hemorrhagic shock [53] and LPS-induced sepsis [54] resulting in overall better survival rates compared with etomidate parent compound. Recently completed studies in rats and beagle dogs have provided preclinical evidence about ET-26HCl safety profile [55,56,57], thus setting the stage for further testing in clinical trials.

The controversial findings and surprisingly few studies describing the effects of etomidate on the developing brain necessitate further investigation. Although earlier studies reported etomidate to be seemingly innocuous to immature brains, a recent study highlighted the proapoptotic effects of etomidate on hippocampal neurons in adult rats [58]. The development of novel etomidate analogs with fewer side effects, such as CPMM and ET-26HCl might reignite the interest and trigger new research of its potential applicability in pediatric anesthesia.

QUATERNARY LIDOCAINE DERIVATIVES: NOVEL DEVELOPMENTS IN REGIONAL ANESTHESIA

In contrast to the recent trends in general anesthetic development where the focus is on quick ‘on’ and ‘off’ effects and rapid metabolism, which allows for greater anesthesia control and easier titration, recent advances in the development of local anesthetics have focused on increasing the duration of action while keeping the systemic adverse effects to a minimum [3]. The short duration of action remains the major shortcoming of lidocaine, limiting its use in certain clinical scenarios. Although the discovery of bupivacaine seemed to solve this problem, the pronounced cardiotoxicity with difficult resuscitation remains a concern in the clinical setting [59■■].

A novel class of local anesthetics known as quaternary lidocaine derivatives (QLD) has recently gained significant research interest with high hopes that they might solve some of the issues described with current local anesthetics [59■■]. Although the mechanism of action of QLD is identical to other local anesthetics [60], the hydrophilic nature and permanent positive charge requires substantial time and concentration gradient to penetrate the neuronal and axonal membrane, which accounts for both the slow onset and prolonged duration of action of QLD.

QX-314, the QLD prototype, applied in three different animal models produced between 6-fold and 12-fold longer duration of anesthesia compared with equimolar lidocaine concentration [61]. Unfortunately, the significant tissue inflammation and systemic toxicity associated with QX-314 [6264] precluded its clinical applicability. The substitution of one hydrogen group (-H) for hydroxyl group (-OH) yielded the newest QLD compound named QX-OH. When three concentrations of QX-OH were compared with equimolar QX-314 and bupivacaine using well established in vivo models, it was shown that QX-OH provided even longer duration of anesthesia and significantly less inflammation; however, because of the permanent charge and hydrophilicity of QX-OH, the rate of onset remains a major issue [65].

Ultimately, it was suggested that transient receptor potential vanilloid 1 (TRPV1) receptors could provide an aqueous route for QLD entry into the neurons [59■■]. As local anesthetics are known to open TRPV1 receptors [66,67], combining QX-OH with levobupivacaine not only dramatically shortened the latency of local anesthetic effect but also prolonged its duration compared with QX-OH alone with minimal local tissue inflammation [68,69]. Furthermore, the preferential location of TRPV1 on nociceptor neurons might provide better pain control with less motor nerve paralysis [70].

Regional anesthesia provides better hemodynamic and respiratory stability with fewer gastrointestinal complications compared with general anesthesia. Although recent evidence suggests that lidocaine causes significant neuroapoptosis both in vivo and in vitro [71], it remains unclear whether local anesthetics are damaging to the developing neurons [20,72]. Nonetheless, the permanent charge of QLD and increased hydrophilicity of QX-OH could help prevent theblood–brain barrier crossing of these novel drugs, and therefore, may minimize the neurotoxic potential. For this reason, further studies are needed to establish whether QLD-based regional anesthesia could serve as a valuable alternative to general anesthesia in childhood, whenever applicable.

XENON: AN ANESTHETIC-SPARING AGENT

Although xenon has been known for its anesthetic properties for some years now, it has not received much attention as an anesthetic for widespread use [73]. In addition to excellent cardiovascular stability [73], preclinical evidence suggests xenon might be neuroprotective especially when used as an adjuvant to other commonly used anesthetics [7477].

Recent warnings about clinical use of anesthetics in young children issued by Food and Drud Administration (FDA) [78] and xenon’s ability to protect against general anesthetics’ developmental neurotoxicity [7477] brought its clinical potential to the forefront. The feasibility of combined xenon and sevoflurane anesthesia was first demonstrated in 2017 on children under the age of 4 undergoing cardiac catheterization [79]. Along with the satisfactory depth of anesthesia, the addition of xenon decreased the requirement for vasopressors and improved cerebral oxygenation. In the latest study comparing the effects of xenon and sevoflurane anesthesia on postoperative neural injury biomarkers, xenon was at least as effective as sevoflurane but the neuroprotective properties could not be confirmed possibly because of overall low incidence of postoperative cognitive dysfunction in the study group [80■■].

Although the xenon production is especially costly [73], the favorable cardiovascular profile and potential neuroprotective properties could mean that the use of xenon as an adjuvant anesthetic might increase in the upcoming years. Although further human studies are needed, the technical feasibility of xenon anesthesia and substantial preclinical evidence might bring this almost forgotten anesthetic back in the operating rooms.

NEUROACTIVE STEROIDS: ANESTHESIA REVISITED FOR THE 21ST CENTURY

Almost 50 years ago, Gyermek and Soyka Lester [81] published an extensive review of a contemporary anesthetic, Alphadione (Althesin), which was a mixture of neuroactive steroid alphaxalone and its 21-acetoxy ester and concluded that alphaxalone, the carrier of the anesthetic effect, was a nearly ideal general anesthetic. In addition to rapid onset of action, alphaxalone had impressively high therapeutic index and hypnotic potency, offered good cardiovascular and respiratory stability, minimal cumulative effects, no injection-site injuries and few drug interactions [81]. Unfortunately, the Althesin formulation contained Cremophor EL, which was likely the culprit for unacceptably high rate of anaphylactic reactions associated with its use [82] leading to its withdrawal from the market in 1984.

The interest towards alphaxalone has recently reemerged and a new formulation with 7-sulfobutyl-ether-β-cyclodextrin as the vehicle was formulated under the name Phaxan. One preclinical study sought to compare the pharmacological properties of Phaxan versus Althesin and propofol in rats [83]. In addition to comparable anesthetic potency and rapid onset and offset of anesthesia, Phaxan had significantly less adverse reactions and higher therapeutic index compared with the other two drugs tested in this study. Alongside similar findings of hypnotic potency and fewer side effects when compared with propofol in humans [84,85], these findings suggest that alphaxalone might, once again, find its place in clinical setting.

We have recently published an extensive summary on mechanism of action of neuroactive steroids and the lack of observable neuronal injury even after prolonged exposures in young animals [86], thus making them a potentially viable alternative to currently used general anesthetics [20]. Briefly, alphaxalone and similar neuroactive steroids, such as investigational drugs, CDNC24 [(3α,5α)-3-hydroxy-13,24-cyclo-18,21-dinorchol-22-en-24-ol) and 3β-OH ((3β,5β,17β)-3-hydroxyandrostane-17-carbonitrile], unlike propofol or ketamine, lack the neuroapoptogenic properties in rat pups [87,88]. Hence, the novel formulation of alphaxalone devoid of its anaphylactogenic properties might be a valuable addition to the anesthetic drug arsenal for children undergoing general anesthesia.

Published studies on the novel anesthetics discussed in this review are summarized in Table 1.

Table 1.

Summary of published clinical and preclinical studies of novel general and local anesthetics

Study type Study Study group Regimen Comparator Endpoints Conclusions
Remimazolam
 Clinical Doi et al., 2020 [24■■] More than 20 years old, less than 100 kg ASA-PS I and II, n = 375 6–12mg/kg/h induction, i.v., 1 mg/kg/h maintenance, i.v. Propofol Efficacy, time to LoC, recovery Well tolerated, noninferior
Doi et al., 2020 [25■■] More than 20 years old; more than 100 kg ASA-PS >III, n = 67 6–12 mg/kg/h induction, i.v. 1–2 mg/kg/h maintenance, i.v. None Efficacy, time to LoC, recovery, control of anesthesia Safe and efficacious
Liu et al., 2021 [26] 35–65 years old SAVR NYHA II or III, n = 60 1.8 mg/kg/h, i.v. Propofol Induction only, ΔHR, ΔMAP, vasopressor use, BIS, Lac, Glu ΔMAP lower in remimazolam, less need for vasopressors
Nakanishi et al., 2021 [27] More than 65 years old TAVR or SAVR severe aortic stenosis ASA-PS IV, n = 20 6 mg/kg/h None vasopressor use, ΔHR, ΔMAP, BIS, time to LoC, time to intubation safe and efficacious with timely vasopressor use
Pastis et al., 2019 [28] More than 18 years old bronchoscopy ASA-PS I, II, III, n = 420 5 mg, i.v. bolus Midazolam, placebo Procedural success, latency to onset and recovery, safety parameters Safe and efficacious, rapid onset and recovery
Borkett et al., 2015 [30] 18–65 years old upper GI endoscopy 60–120 kg, BMI 18–29 ASA-PS I or II n = 100 0.10, 0.15 or 0.20 mg/kg Midazolam Procedural success, latency to onset and recovery, safety parameters Safe and efficacious, rapid onset and recovery at all doses tested
Zhang et al., 2022 [31] Female, 18–55 years old hysteroscopy BMI 18–30 ASA-PS I or II, n = 90 0.25 mg/kg i.v. bolus, 0.48 or 0.6 mg/kg/h, i.V. Propofol Procedural success, latency to onset and recovery, safety parameters Safe and efficacious, less adverse effects
Tan et al., 2022 [40] More than 60 years old, upper GI endoscopy ASA-PS I or II, n = 99 0.1 or 0.2 mg/kg Propofol Cognitive outcomes in elderly patients, success rates, onset and recovery Cognitive impairment, 0.2>0.1 mg/kg less hypotension in 0.1 mg/kg
Horikoshi et al, 2021 [42] 4-year-old boy Duchenne muscular dystrophy, inguinal herniorrhaphy 3 mg i.v. bolus induction, 15 mg/h i.v. maintenance None Efficacy, feasibility, recovery Safe and effective
 Preclinical Liu et al., 2021 [32■■] Mice, 8 weeks old 20 mg/kg LPS primary BMDM 16 mg/kg i.p., 30 min before LPS 8 mg/kg i.p., 30 min after LPS None IHC, IF, ELISA, qPCR, WB, flow cytometry ↓LPS-induced cell death ↓TNFα, IL-1β, IL-6 ↓tissue inflammation and necrosis ↓TLR4, NF-kB signaling ↓pAkt, ↓pERKl/2
Xu et al., 2021 [33] Primary astrocytes, U-11 8MG and U87MG glioma cell lines 2–20 μmol/l RFMSP (remimazolam derivative) None Viability, IF, WB, flow cytometry ↓glioma cell viability ↑glioma apoptosis (caspase-3) ↓NF-kB, ↓survivin ↓Bcl-2 and ↑Bax
Xie et al., 2021 [34] Rats, adult CFA 20 μl paw BV2 microglial cells LPS 1 mg/ml 2.5 mg/kg in vivo 200 μg/ml in vitro None ELISA, WB, TEM, IF ↓bradykinin receptor ↓pain and Inflammation ↓NF-kB, ↓autophagy
Kops et al., 2021 [41] Cynomolgus monkeys, 3 years old 5–50 mg/kg/h, i.v. Midazolam, propofol Sedation behavior Rapid and potent sedation dose-dependent synergism with remifentanil
CPMM
 Clinical Valk et al., 2018 [47] 18–45 years old, healthy nonsmoking, ASA-PS I, n = 40 30–60 μg/kg/min for 30 min Propofol, placebo Safety and efficacy, hemodynamic and adrenal Rapid onset and recovery, mild ↑HR and ↑MAP dose-dependent IMM
Struys et al., 2017 [48] 18–45 years old, healthy nonsmoking, BMI 17.5–30, ASA-PS I or II, n = 60 0.03–1 mg/kg, i.v. bolus Placebo Safety and efficacy, dose optimization, onset and recovery Rapid onset and recovery, dose-dependent IMM
 Preclinical Santer et al., 2015 [46] Rats, adult 1 mg/kg LPS 71 mg/kg total, 1 h i.v. infusion Etomidate ELISA (corticosterone and cytokines) Less adrenal suppresion than etomidate ↓IL-1β, IL-6, IL-10 ↓mortality
Valk et al., 2019 [49] Beagle dogs, 8–9 months, n = 14 6 mg/kg i.v. bolus induction 1, 1.5 or 2.3 mg/kg/min maintenance for 2 h None GABAA electrophysiology and toxicology IMM upon use not of ictal origin GABAA and glycin mediated
ET-26HCI
 Preclinical Wang et al., 2017 [50] Rats, adult 1.3–4.5 mg/kg, i.v. Etomidate, propofol ELISA, behavior, pharmacokinetics, onset and recovery Rapid onset and recovery less adrenal suppression than etomidate less ΔMAP than propofol IMM with use
Yang et al., 2017 [51] Beagle dogs, adult Dose range not specified Etomidate, CPMM Onset and recovery, pharmacokinetics, pharmacodynamics Less adrenal suppression than etomidate
Liu et al., 2017 [52] Rats, adult beagle dogs, adult Langendorff rat heart 1, 2, 4 × ED50 Etomidate echocardiography, ECG, electrophysiology ↑myocardial performance similar to etomidate
Wang et al., 2017 [53] Rats, adult uncontrolled hemorrhagic shock model 1.33–3 mg/kg, i.v. bolus Etomidate, propofol Pharmacokinetics, pharmacodynamics, hemodynamics ↑hemodynamic stability ↓lactate
Wang et al, 2017 [54] Rats, adult 1 mg/kg LPS 2 × ED50, i.v. bolus Etomidate, saline IHC (lungs, kidneys) ELISA (plasma cytokines, corticosterone) ↑survival compared to etomidate less adrenal suppression ↓IL-1β, ↓IL-6
Zhang et al., 2021 [55] Rats, adult beagle dogs, 1–2 years old 4, 8, 16 mg/kg, rat, i.v. 8, 12, 16 mg/kg, dog, i.v. Vehicle (propylene glycol) Cardiovascular, respiratory, central nervous system function Mild and reversible adverse effects
Zhang et al., 2020 [56] Beagle dogs, 6–8 months 4–16 mg/kg, i.v., single 8–16 mg/kg, i.v., repeated for 14 days Vehicle (propylene glycol) Toxicology: heart, blood, vital organs No observable side effects at any timepoint
Zhang et al., 2020 [57] Rats, adult 8, 16, 20 mg/kg, i.v., single 8, 12, 16 mg/kg, i.v., repeated for 14 days Vehicle (propylene glycol) Toxicology: urine, blood, vital organs ↑Mortality with repeated doses no serious adverse effects with single doses
QX-314
 Preclinical Lim et al., 2007 [61] Mice, adult, n = 45 guinea pigs, adult, n = 8 10, 30, 70 mmol/l Lidocaine, saline Intradermal (guinea pigs) tail flick and sciatic nerve block (mice) ↑↑Duration slow onset
Shankarappa et al, 2012 [62] Rats, adult males 25 mmol/l QX-222 Sciatic nerve block IHC (myotoxicity) ↑↑Duration severe myotoxicity
Schwarz et al, 2010 [63] Mice, adult female 0.5–10 mmol/l, it Saline Efficacy (lumbosacral block) toxicity (irritation and death) ↑↑Duration severe adverse effects
Cheung et al, 2011 [64] Mice, adult 7.5–30 mg/kg, i.v. Lidocaine Toxic and lethal dose CNS and cardiac toxicity Severe cardiac and CNS toxicity
QX-OH
 Preclinical Zhang et al, 2017 [65] Rats, adult 25, 35, 45 mmol/l QX-314 bupivacaine saline IHC (local inflammation) sciatic nerve block cutaneous trunci pinprick ↑↑↑Duration compared with QX-314 and bupivacaine less local toxicity slow onset
Zhao et al, 2018 [68] Rats, adult mice, adult total knee arthroplasty 35 mmol/l + 10 mmol/l levobupivacaine Liposome bupivacaine, saline IHC (local inflammation) behavior (open field, thermal nociception, autonomic activity) ↑↑↑Duration of block minimal local toxicity when combined
Yin et al, 2019 [69] Rats, adult subcutaneous infiltration sciatic nerve block 35 mmol/l + 10 mmol/l levobupivacaine QX-OH levobupivacaine saline Onset and recovery duration of effect local and systemic toxicity Faster onset ↑↑↑duration ↓local and systemic toxicity when combined
Xenon
 Clinical Devroe et al, 2017 [79] Less than 4 years old elective cardiac catheterization n = 40 50–65% + sevoflurane Sevoflurane Hemodynamic stability feasibility, efficacy and recovery safety Safe and efficacious ↓vasopressor use
McGuigan et al., 2022 [80■■] More than 50 years old ESWL, ASA-PS I and II n = 23 60% end-tidal, 6 ml/kg Sevoflurane Neural injury biomarkers cognitive assessment No change in injury biomarkers no difference in cognitive assessment
 Preclinical Cattano et al., 2008 [74] Mice, 7 days old 70%±0.75% isoflurane for 4 h Isoflurane IHC (neuroapoptosis) ↓Caspase-3 in caudate/putamen when combined with isoflurane
Gill et al., 2020 [75] Rats, 8 days old 35% + 1.8% sevoflurane 70% + 0.9% sevoflurane for 6 h Sevoflurane IHC (neuroapoptosis) ↓Caspase-3 in cortex and caudate/putamen (70% only)
Ma et al., 2007 [76] Rats, 7 days old 30, 60, 75%±0.75% isoflurane for 6 h Isoflurane IHC and WB (neuroapoptosis) ↓Caspase-3, caspase-9 ↓cytochrome c
Shu et al., 2010 [77] Rats, 7 days old 70%, pretreatment for 2 h Nitrous oxide and isoflurane IHC and WB (neuroapoptosis) fear conditioning (memory) ↓Neuroapoptosis in cortex and hippocampus ↑memory
Neuroactive steroids
 Clinical Monagle et al., 2015 [84] 18–40 years old males BMI 18–25, ASA-PS I, healthy volunteers n = 24 0.42–0.55 mg/kg (Phaxan) dose titration to BIS<50 Propofol Onset and recovery cardiovascular and respiratory effects adverse effects cognitive function Rapid onset and recovery ↓cardiovascular and respiratory depression ↓adverse effects normal cognitive performance
 Preclinical Goodchild et al., 2015 [83] Rats, adult 1.2–15 mg/kg, i.v. (Phaxan) Propofol althesin Effective and lethal dose cardiovascular effects lethality Rapid onset and recovery equipotent to Althesin higher therapeutic index less cardiovascular depression
Atluri et al., 2018 [87] Rats, 7 days old 5–10 mg/kg, i.p. every 2 h for 12 h (3β-OH) Ketamine Effective and lethal dose, IHC (neuroapoptosis) behavior (radial arm maze) electrophysiology ↓Neuroapoptosis subiculum, thalamus and cortex ↑memory T-type calcium channel block, ↓spontaneous GABA release
Tesic et al., 2020 [88] Rats, 7 days old 10 mg/kg, i.p. every hour for 6h (alphaxalone and CDNC24) Propofol Effective and lethal dose, IHC (neuroapoptosis) electrophysiology ↓Neuroapoptosis subiculum and medial prefrontal cortex GABA-potentiating effect

ASA-PS, American Society of Anesthesiologists Physical Status; BIS, bispectral index; BMDM, bone marrow-derived macrophages; CFA, complete Freund’s adjuvant; CNS, central nervous system; ESWL, extracorporeal shock wave lithotripsy; GABA, gamma-amino butyric acid; GI, gastrointestinal; Glu, glucose; HR, heart rate; IF, immunofluorescence; IHC, immunohistochemistry; IL, interleukin; IMM, involuntary muscle movement; i.p., intraperitoneally; i.v., intravenously; Lac, lactose; LoC, loss of consciousness; LPS, lipopolysaccharide; MAP, mean arterial pressure; NF-kB, nuclear factor kappa b; NYHA, New York Heart association; qPCR, quantitative PCR; SAVR, surgical aortic valve replacement; TAVR, transcatheter aortic valve replacement; TEM, transmission electron microscopy; TLR4, toll-like receptor 4; TNFα, tumor necrosis factor alpha; WB, western blot.

CONCLUSION

Despite established preclinical and growing clinical evidence indicating the neurotoxic properties of virtually all commonly used general anesthetics with long-lasting consequences on normal behavioral development, the best path forward remains unclear. As the evidence in this field continues to mount (over 1000 publications around the world in a variety of animal species – from nematodes to nonhuman primates and humans), we have seen an interesting spectrum of corollaries ranging from minimizing it, to being sceptical but willing to consider it, to actually contemplating the phenomenon as important and relevant. Although the field is pondering the best course of action, we believe the need for the development of new and safer anesthetics cannot be underestimated especially in view of the fact that many therapeutic interventions have tried and failed to conclusively convey any neuroprotection against anesthesia-induced developmental neurotoxicity [20], leaving us perplexed as to how to proceed. Perhaps it is time to question the entrenched fondness for our current anesthetics and embrace the new pharmacotherapeutics for the sake of the youngest, sickest and most vulnerable members of our community.

KEY POINTS.

  • Remimazolam is a ‘soft’ analog of midazolam with safety and applicability comparable to propofol causing benzodiazepine-based anesthesia to be revisited.

  • Two of the etomidate analogs, CPMM and ET-26HCl, are currently being investigated as intravenous anesthetics with less adrenocortical suppression while maintaining hemodynamic stability.

  • Quaternary lidocaine derivatives are being developed as more potent, longer lasting regional anesthetics with unique pharmacological features.

  • Xenon and alphaxalone are being revisited as potential general anesthetics in humans with potentially important safety benefits in children.

  • Further preclinical evidence is necessary to establish the neurotoxic profile of these novel anesthetic compounds.

Financial support and sponsorship

Supported in part by funds from the Department of Anesthesiology at the University of Colorado Anschutz Medical Campus, CU Medicine Endowment (to V.J.-T.), NIH R01 GM123746 (to V.J.-T.), NIH R01 HD097990 (V.J.-T.), NIH R01 HD044517 (V.J.-T.), NIH R01 HD044517-S (V.J.-T.), R01 GM118197 (V.J.-T.) and R01 GM118197-S1 (V.J.-T.).

Footnotes

Conflicts of interest

There are no conflicts of interest.

REFERENCES AND RECOMMENDED READING

Papers of particular interest, published within the annual period of review, have been highlighted as:

■ of special interest

■■ of outstanding interest

  • 1.Johnson KB. New horizons in sedative hypnotic drug development: fast, clean, and soft. Anesth Analg 2012; 115:220–222. [DOI] [PubMed] [Google Scholar]
  • 2.Sneyd JR, Rigby-Jones AE. New drugs and technologies, intravenous anaesthesia is on the move (again). Br J Anaesth 2010; 105:246–254. [DOI] [PubMed] [Google Scholar]
  • 3.Macfarlane AJR, Gitman M, Bornstein KJ, et al. Updates in our understanding of local anaesthetic systemic toxicity: a narrative review. Anaesthesia 2021; 76(Suppl 1):27–39. [DOI] [PubMed] [Google Scholar]
  • 4.Jevtovic-Todorovic V, Hartman RE, Izumi Y, et al. Early exposure to common anesthetic agents causes widespread neurodegeneration in the developing rat brain and persistent learning deficits. J Neurosci 2003; 23:876–882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Rizzi S, Carter LB, Ori C, Jevtovic-Todorovic V. Clinical anesthesia causes permanent damage to the fetal guinea pig brain. Brain Pathol 2008; 18:198–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Slikker W Jr, Zou X, Hotchkiss CE, et al. Ketamine-induced neuronal cell death in the perinatal rhesus monkey. Toxicol Sci 2007; 98:145–158. [DOI] [PubMed] [Google Scholar]
  • 7.Paule MG, Li M, Allen RR, et al. Ketamine anesthesia during the first week of life can cause long-lasting cognitive deficits in rhesus monkeys. Neurotoxicol Teratol 2011; 33:220–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Schenning KJ, Noguchi KK, Martin LD, et al. Isoflurane exposure leads to apoptosis of neurons and oligodendrocytes in 20- and 40-day old rhesus macaques. Neurotoxicol Teratol 2017; 60:63–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Noguchi KK, Johnson SA, Dissen GA, et al. Isoflurane exposure for three hours triggers apoptotic cell death in neonatal macaque brain. Br J Anaesth 2017; 119:524–531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ye Z, Li Q, Guo Q, et al. Ketamine induces hippocampal apoptosis through a mechanism associated with the caspase-1 dependent pyroptosis. Neuropharmacology 2018; 128:63–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Olney JW, Tenkova T, Dikranian K, et al. Ethanol-induced caspase-3 activation in the in vivo developing mouse brain. Neurobiol Dis 2002; 9:205–219. [DOI] [PubMed] [Google Scholar]
  • 12.Raper J, Alvarado MC, Murphy KL, Baxter MG. Multiple anesthetic exposure in infant monkeys alters emotional reactivity to an acute stressor. Anesthesiology 2015; 123:1084–1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Creeley C, Dikranian K, Dissen G, et al. Propofol-induced apoptosis of neurones and oligodendrocytes in fetal and neonatal rhesus macaque brain. Br J Anaesth 2013; 110(Suppl 1):i29–i38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wilder RT, Flick RP, Sprung J, et al. Early exposure to anesthesia and learning disabilities in a population-based birth cohort. Anesthesiology 2009; 110: 796–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hu D, Flick RP, Zaccariello MJ, et al. Association between exposure of young children to procedures requiring general anesthesia and learning and behavioral outcomes in a population-based birth cohort. Anesthesiology 2017; 127:227–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ing C, DiMaggio C, Whitehouse A, et al. Long-term differences in language and cognitive function after childhood exposure to anesthesia. Pediatrics 2012; 130:e476–e485. [DOI] [PubMed] [Google Scholar]
  • 17.DiMaggio C, Sun LS, Li G. Early childhood exposure to anesthesia and risk of developmental and behavioral disorders in a sibling birth cohort. Anesth Analg 2011; 113:1143–1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ing C, Hegarty MK, Perkins JW, et al. Duration of general anaesthetic exposure in early childhood and long-term language and cognitive ability. Br J Anaesth 2017; 119:532–540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Flick RP,Katusic SK,Colligan RC,et al. Cognitive and behavioraloutcomesafter earlyexposuretoanesthesiaandsurgery. Pediatrics 2011;128:e1053–e1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Useinovic N, Maksimovic S, Near M, et al. Do we have viable protective strategies against anesthesia-induced developmental neurotoxicity? Int J Mol Sci 2022; 23:1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Russell IF. Midazolam-alfentanil: an anaesthetic? An investigation using the isolated forearm technique. Br J Anaesth 1993; 70:42–46. [DOI] [PubMed] [Google Scholar]
  • 22.Schwender D, Golling W, Klasing S, et al. Effects of surgical stimulation on midlatency auditory evoked potentials during general anaesthesia with propofol/fentanyl, isoflurane/fentanyl and flunitrazepam/fentanyl. Anaesthesia 1994; 49:572–578. [DOI] [PubMed] [Google Scholar]
  • 23.Kilpatrick GJ, McIntyre MS, Cox RF, et al. CNS 7056: a novel ultra-shortacting benzodiazepine. Anesthesiology 2007; 107:60–66. [DOI] [PubMed] [Google Scholar]
  • 24.■■.Doi M, Morita K, Takeda J, et al. Efficacy and safety of remimazolam versus propofol for general anesthesia: a multicenter, single-blind, randomized, parallel-group, phase IIb/III trial. J Anesth 2020; 34:543–553. [DOI] [PubMed] [Google Scholar]; A large multicenter clinical trial demonstrating the general anesthesia induction and maintenance with remimazolam versus propofol in low-risk patients.
  • 25.■■.Doi M, Hirata N, Suzuki T, et al. Safety and efficacy of remimazolam in induction and maintenance of general anesthesia in high-risk surgical patients (ASA Class III): results of a multicenter, randomized, double-blind, parallel group comparative trial. J Anesth 2020; 34:491–501. [DOI] [PubMed] [Google Scholar]; A particularly interesting multicenter clinical trial on remimazolam induction and maintenance of general anesthesia in high-risk patients, showing excellent safety and tolerability.
  • 26.Liu T, Lai T, Chen J, et al. Effect of remimazolam induction on hemodynamics in patients undergoing valve replacement surgery: a randomized, double-blind, controlled trial. Pharmacol Res Perspect 2021; 9:e00851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.■.Nakanishi T, Sento Y, Kamimura Y, et al. Remimazolam for induction of anesthesia in elderly patients with severe aortic stenosis: a prospective, observational pilot study. BMC Anesthesiol 2021; 21:306. [DOI] [PMC free article] [PubMed] [Google Scholar]; A clinical study, which highlights the hemodynamic stability of anesthesia induction with remimazolam in patients with reduced cardiovascular reserve.
  • 28.Pastis NJ, Yarmus LB, Schippers F, et al. Safety and efficacy of remimazolam compared with placebo and midazolam for moderate sedation during bronchoscopy. Chest 2019; 155:137–146. [DOI] [PubMed] [Google Scholar]
  • 29.Rex DK, Bhandari R, Desta T, et al. A phase III study evaluating the efficacy and safety of remimazolam (CNS 7056) compared with placebo and midazolam in patients undergoing colonoscopy. Gastrointest Endosc 2018; 88:427–437; e426. [DOI] [PubMed] [Google Scholar]
  • 30.Borkett KM, Riff DS, Schwartz HI, et al. A phase IIa, randomized, double-blind study of remimazolam (CNS 7056) versus midazolam for sedation in upper gastrointestinal endoscopy. Anesth Analg 2015; 120:771–780. [DOI] [PubMed] [Google Scholar]
  • 31.■.Zhang S, Wang J, Ran R, et al. Efficacy and safety of remimazolam tosylate in hysteroscopy: a randomized, single-blind, parallel controlled trial. J Clin Pharm Ther 2022; 47:55–60. [DOI] [PubMed] [Google Scholar]; A recent study demonstrating safety and efficacy of remimazolam for procedural sedation versus standard propofol-based sedation, showing less adverse effects associated with remimazolam.
  • 32.■■.Liu X, Lin S, Zhong Y, et al. Remimazolam protects against LPS-induced endotoxicity improving survival of endotoxemia mice. Front Pharmacol 2021; 12:739603. [DOI] [PMC free article] [PubMed] [Google Scholar]; Preclinical study describing the molecular underpinnings of signaling interference of remimazolam with lipopolysaccharide, highlighting the toll-like receptors, NF-kB and downstream signaling molecules as potential targets modulated by remimazolam.
  • 33.Xu W, Hu J, Liu W, et al. Remimazolan inhibits glioma cell growth and induces apoptosis through down-regulation of NF-κB pathway. IUBMB Life 2021; 73:341–348. [DOI] [PubMed] [Google Scholar]
  • 34.Xie H, Lu F, Liu W, et al. Remimazolam alleviates neuropathic pain via regulating bradykinin receptor B1 and autophagy. J Pharm Pharmacol 2021; 73:1643–1651. [DOI] [PubMed] [Google Scholar]
  • 35.Yon JH, Daniel-Johnson J, Carter LB, Jevtovic-Todorovic V. Anesthesia induces neuronal cell death in the developing rat brain via the intrinsic and extrinsic apoptotic pathways. Neuroscience 2005; 135:815–827. [DOI] [PubMed] [Google Scholar]
  • 36.Yon JH, Carter LB, Reiter RJ, Jevtovic-Todorovic V. Melatonin reduces the severity of anesthesia-induced apoptotic neurodegeneration in the developing rat brain. Neurobiol Dis 2006; 21:522–530. [DOI] [PubMed] [Google Scholar]
  • 37.Boscolo A, Milanovic D, Starr JA, et al. Early exposure to general anesthesia disturbs mitochondrial fission and fusion in the developing rat brain. Anesthesiology 2013; 118:1086–1097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Boscolo A, Starr JA, Sanchez V, et al. The abolishment of anesthesia-induced cognitive impairment by timely protection of mitochondria in the developing rat brain: the importance of free oxygen radicals and mitochondrial integrity. Neurobiol Dis 2012; 45:1031–1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lu LX, Yon JH, Carter LB, Jevtovic-Todorovic V. General anesthesia activates BDNF-dependent neuroapoptosis in the developing rat brain. Apoptosis 2006; 11:1603–1615. [DOI] [PubMed] [Google Scholar]
  • 40.■.Tan Y, Ouyang W, Tang Y, et al. Effect of remimazolam tosilate on early cognitive function in elderly patients undergoing upper gastrointestinal endoscopy. J Gastroenterol Hepatol 2022; 37:576–583. [DOI] [PMC free article] [PubMed] [Google Scholar]; A clinical trial including elderly patients undergoing procedural sedation with two different doses of remimazolam, with concerning findings regarding the cognitive impairments associated with higher dose of remimazolam.
  • 41.Kops MS, Pesic M, Petersen KU, et al. Impact of concurrent remifentanil on the sedative effects of remimazolam, midazolam and propofol in cynomolgus monkeys. Eur J Pharmacol 2021; 890:173639. [DOI] [PubMed] [Google Scholar]
  • 42.■.Horikoshi Y, Kuratani N, Tateno K, et al. Anesthetic management with remimazolam for a pediatric patient with Duchenne muscular dystrophy. Medicine (Baltimore) 2021; 100:e28209. [DOI] [PMC free article] [PubMed] [Google Scholar]; A case report highlighting the uneventful inguinal herniorrhaphy under general anesthesia induced and maintained with remimazolam in a 4-year-old boy with Duchenne muscular dystrophy.
  • 43.Ledingham IM, Watt I. Influence of sedation on mortality in critically ill multiple trauma patients. Lancet 1983; 1:1270. [DOI] [PubMed] [Google Scholar]
  • 44.Wagner RL, White PF, Kan PB, et al. Inhibition of adrenal steroidogenesis by the anesthetic etomidate. N Engl J Med 1984; 310:1415–1421. [DOI] [PubMed] [Google Scholar]
  • 45.Hildreth AN, Mejia VA, Maxwell RA, et al. Adrenal suppression following a single dose of etomidate for rapid sequence induction: a prospective randomized study. J Trauma 2008; 65:573–579. [DOI] [PubMed] [Google Scholar]
  • 46.Santer P, Pejo E, Feng Y, et al. Cyclopropyl-methoxycarbonyl metomidate: studies in a lipopolysaccharide inflammatory model of sepsis. Anesthesiology 2015; 123:368–376. [DOI] [PubMed] [Google Scholar]
  • 47.Valk BI, Absalom AR, Meyer P, et al. Safety and clinical effect of i.v. infusion of cyclopropyl-methoxycarbonyl etomidate (ABP-700), a soft analogue of etomidate, in healthy subjects. Br J Anaesth 2018; 120:1401–1411. [DOI] [PubMed] [Google Scholar]
  • 48.Struys M, Valk BI, Eleveld DJ, et al. A phase 1, single-center, double-blind, placebo-controlled study in healthy subjects to assess the safety, tolerability, clinical effects, and pharmacokinetics-pharmacodynamics of intravenous cyclopropyl-methoxycarbonylmetomidate (ABP-700) after a single ascending bolus dose. Anesthesiology 2017; 127:20–35. [DOI] [PubMed] [Google Scholar]
  • 49.Valk BI, McGrath M, Lehoux D, et al. Toxicologic and inhibitory receptor actions of the etomidate analog ABP-700 and its metabolite CPM-acid. Anesthesiology 2019; 131:287–304. [DOI] [PubMed] [Google Scholar]
  • 50.Wang B, Yang J, Chen J, et al. An etomidate analogue with less adrenocortical suppression, stable hemodynamics, and improved behavioral recovery in rats. Anesth Analg 2017; 125:442–450. [DOI] [PubMed] [Google Scholar]
  • 51.Yang J, Kang Y, Wang B, et al. Metabolite-inactive etomidate analogues alleviating suppression on adrenal function in Beagle dogs. Eur J Pharm Sci 2017; 99:343–349. [DOI] [PubMed] [Google Scholar]
  • 52.Liu X, Song H, Yang J, et al. The etomidate analog ET-26 HCl retains superior myocardial performance: comparisons with etomidate in vivo and in vitro. PLoS One 2018; 13:e0190994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Wang B, Chen S, Yang J, et al. ET-26 hydrochloride (ET-26 HCl) has similar hemodynamic stability to that of etomidate in normal and uncontrolled hemorrhagic shock (UHS) rats. PLoS One 2017; 12:e0183439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wang B, Jiang J, Yang J, et al. Pharmacologic studies on ET-26 hydrochloride in a rat model of lipopolysaccharide-induced sepsis. Eur J Pharm Sci 2017; 109:441–445. [DOI] [PubMed] [Google Scholar]
  • 55.■.Zhang Y, Jiang Y, Chang P, et al. Safety pharmacology study of ET-26 hydrochloride, a potential drug for intravenous general anesthesia, in rats and beagle dogs. Front Pharmacol 2021; 12:679381. [DOI] [PMC free article] [PubMed] [Google Scholar]; An extensive pharmacology and toxicology report with regard to use of ET26-HCl, a novel etomidate analog, in rats and beagle dogs, setting the stage for future clinical trials.
  • 56.Zhang Y, Deng C, Gong D, et al. Preclinical safety evaluation of ET-26 hydrochloride, a novel intravenous anesthetic agent, in beagle dogs. J Appl Toxicol 2020; 40:679–690. [DOI] [PubMed] [Google Scholar]
  • 57.Zhang Y, Zhang Y, Deng C, et al. Nonclinical single- and repeated-dose toxicity studies of ET-26 hydrochloride in rats. J Appl Toxicol 2020; 40:1099–1112. [DOI] [PubMed] [Google Scholar]
  • 58.■.Alipour M, Asl MK, Khordad E, et al. Histopathological study on neuroapop-totic alterations induced by etomidate in rat hippocampus. Acta Histochem 2021; 123:151693. [DOI] [PubMed] [Google Scholar]; In contrast with the earlier reports with regard to etomidate application in neonatal animals, this study found increased hippocampal neuroapoptosis in adult rats following etomidate administration.
  • 59.■■.Wang Q, Zhang Y, Liu J, Zhang W. Quaternary lidocaine derivatives: past, present, and future. Drug Des Devel Ther 2021; 15:195–207. [DOI] [PMC free article] [PubMed] [Google Scholar]; A highly informative review explaining the rationale for development of quaternary lidocaine derivatives, with focus on pharmacodynamic and toxicity profiles of QX-314 and QX-OH.
  • 60.Taylor A, McLeod G. Basic pharmacology of local anaesthetics. BJA Educ 2020; 20:34–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Lim TK, Macleod BA, Ries CR, Schwarz SK. The quaternary lidocaine derivative, QX-314, produces long-lasting local anesthesia in animal models in vivo. Anesthesiology 2007; 107:305–311. [DOI] [PubMed] [Google Scholar]
  • 62.Shankarappa SA, Sagie I, Tsui JH, et al. Duration and local toxicity of sciatic nerve blockade with coinjected site 1 sodium-channel blockers and quaternary lidocaine derivatives. Reg Anesth Pain Med 2012; 37:483–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Schwarz SK, Cheung HM, Ries CR, et al. Lumbar intrathecal administration of the quaternary lidocaine derivative, QX-314, produces irritation and death in mice. Anesthesiology 2010; 113:438–444. [DOI] [PubMed] [Google Scholar]
  • 64.Cheung HM, Lee SM, MacLeod BA, et al. A comparison of the systemic toxicity of lidocaine versus its quaternary derivative QX-314 in mice. Can J Anaesth 2011; 58:443–450. [DOI] [PubMed] [Google Scholar]
  • 65.Zhang Y, Yang J, Yin Q, et al. QX-OH, a QX-314 derivative agent, produces long-acting local anesthesia in rats. Eur J Pharm Sci 2017; 105:212–218. [DOI] [PubMed] [Google Scholar]
  • 66.Leffler A, Fischer MJ, Rehner D, et al. The vanilloid receptor TRPV1 is activated and sensitized by local anesthetics in rodent sensory neurons. J Clin Invest 2008; 118:763–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Brenneis C, Kistner K, Puopolo M, et al. Bupivacaine-induced cellular entry of QX-314 and its contribution to differential nerve block. Br J Pharmacol 2014; 171:438–451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zhao W, Yang J, Zhang Y, et al. QX-OH/Levobupivacaine: fixed-dose combination to provide a long-acting postoperative pain of knee surgery in rodents. Eur J Pharm Sci 2018; 111:418–424. [DOI] [PubMed] [Google Scholar]
  • 69.Yin Q, Zhang Y, Lv R, et al. A fixed-dose combination, QXOH/levobupivacaine, produces long-acting local anesthesia in rats without additional toxicity. Front Pharmacol 2019; 10:243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Binshtok AM, Bean BP, Woolf CJ. Inhibition of nociceptors by TRPV1-mediated entry of impermeant sodium channel blockers. Nature 2007; 449:607–610. [DOI] [PubMed] [Google Scholar]
  • 71.Lirk P, Haller I, Myers RR, et al. Mitigation of direct neurotoxic effects of lidocaine and amitriptyline by inhibition of p38 mitogen-activated protein kinase in vitro and in vivo. Anesthesiology 2006; 104:1266–1273. [DOI] [PubMed] [Google Scholar]
  • 72.Hampl K, Steinfeldt T, Wulf H. Spinal anesthesia revisited: toxicity of new and old drugs and compounds. Curr Opin Anaesthesiol 2014; 27:549–555. [DOI] [PubMed] [Google Scholar]
  • 73.Sanders RD, Ma D, Maze M. Xenon: elemental anaesthesia in clinical practice. Br Med Bull 2004; 71:115–135. [DOI] [PubMed] [Google Scholar]
  • 74.Cattano D, Williamson P, Fukui K, et al. Potential of xenon to induce or to protect against neuroapoptosis in the developing mouse brain. Can J Anaesth 2008; 55:429–436. [DOI] [PubMed] [Google Scholar]
  • 75.Gill H, Pickering AE. The effects of xenon on sevoflurane anesthesia-induced acidosis and brain cell apoptosis in immature rats. Paediatr Anaesth 2021; 31:372–374. [DOI] [PubMed] [Google Scholar]
  • 76.Ma D, Williamson P, Januszewski A, et al. Xenon mitigates isoflurane-induced neuronal apoptosis in the developing rodent brain. Anesthesiology 2007; 106:746–753. [DOI] [PubMed] [Google Scholar]
  • 77.Shu Y, Patel SM, Pac-Soo C, et al. Xenon pretreatment attenuates anestheticinduced apoptosis in the developing brain in comparison with nitrous oxide and hypoxia. Anesthesiology 2010; 113:360–368. [DOI] [PubMed] [Google Scholar]
  • 78.Olutoye OA, Baker BW, Belfort MA, Olutoye OO. Food and Drug Administration warning on anesthesia and brain development: implications for obstetric and fetal surgery. Am J Obstet Gynecol 2018; 218:98–102. [DOI] [PubMed] [Google Scholar]
  • 79.Devroe S, Meeusen R, Gewillig M, et al. Xenon as an adjuvant to sevoflurane anesthesia in children younger than 4 years of age, undergoing interventional or diagnostic cardiac catheterization: a randomized controlled clinical trial. Paediatr Anaesth 2017; 27:1210–1219. [DOI] [PubMed] [Google Scholar]
  • 80.■■.McGuigan S, Evered L, Scott DA, et al. Comparing the effect of xenon and sevoflurane anesthesia on postoperative neural injury biomarkers: a randomized controlled trial. Med Gas Res 2022; 12:10–17. [DOI] [PMC free article] [PubMed] [Google Scholar]; Recent study comparing the neural injury biomarkers and postoperative cognitive function after anesthesia with xenon versus sevoflurane, indicating the noninferiority of xenon as a standalone anesthetic in this study group.
  • 81.Gyermek L, Soyka Lester F. Steroid anesthetics. Anesthesiology 1975; 42:331–344. [DOI] [PubMed] [Google Scholar]
  • 82.Radford SG, Lockyer JA, Simpson PJ. Immunological aspects of adverse reactions to althesin. Br J Anaesth 1982; 54:859–864. [DOI] [PubMed] [Google Scholar]
  • 83.Goodchild CS, Serrao JM, Kolosov A, Boyd BJ. Alphaxalone reformulated: a water-soluble intravenous anesthetic preparation in sulfobutyl-ether-β-cyclodextrin. Anesth Analg 2015; 120:1025–1031. [DOI] [PubMed] [Google Scholar]
  • 84.Monagle J, Siu L, Worrell J, et al. A phase 1c trial comparing the efficacy and safety of a new aqueous formulation of alphaxalone with propofol. Anesth Analg 2015; 121:914–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Goodchild CS, Serrao JM, Sear JW, Anderson BJ. Pharmacokinetic and pharmacodynamic analysis of alfaxalone administered as a bolus intravenous injection of phaxan in a phase 1 randomized trial. Anesth Analg 2020; 130: 704–714. [DOI] [PubMed] [Google Scholar]
  • 86.Maksimovic S, Useinovic N, Quillinan N, et al. General anesthesia and the young brain: the importance of novel strategies with alternate mechanisms of action. Int J Mol Sci 2022; 23:. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Atluri N, Joksimovic SM, Oklopcic A, et al. A neurosteroid analogue with T-type calcium channel blocking properties is an effective hypnotic, but is not harmful to neonatal rat brain. Br J Anaesth 2018; 120:768–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.■.Tesic V, Joksimovic SM, Quillinan N, et al. Neuroactive steroids alphaxalone and CDNC24 are effective hypnotics and potentiators of GABA(A) currents, but are not neurotoxic to the developing rat brain. Br J Anaesth 2020; 124:603–613. [DOI] [PMC free article] [PubMed] [Google Scholar]; A histological and electrophysiological report on nonneurotoxicity of two neurosteroids compared vis-à-vis with propofol in neonatal rats.

RESOURCES