Korean J Anesthesiol Search

CLOSE


Korean J Anesthesiol > Volume 76(2); 2023 > Article
Sun, Zhu, Esmaeili, Tang, and Wu: Mechanisms and implications in gene polymorphism mediated diverse reponses to sedatives, analgesics and muscle relaxants

Abstract

Responses to sedatives, analgesics and muscle relaxants vary among patients under general anesthesia, which could be ascribed to the disparities of clinical characteristics and genetic factors of individuals. Accumulating researches have indicated that gene polymorphisms of the receptors, transporters and metabolizing enzymes associated with anesthetics play a considerable role in their efficacy. However, a systematically summarized study on the mechanisms of gene polymorphisms on pharmacodynamics and pharmacokinetics of anesthetics is still lacking. In this paper, the recent researches on pharmacogenomics of sedatives, analgesics and muscle relaxants are comprehensively reviewed, and the contributions and mechanisms of polymorphisms to the differences of individual efficacy of these drugs are discussed, so as to provide guidance for the formulation of a rational anesthesia regimen for patients with various genotypes.

Introduction

Both clinical practices and scientific researches have testified that there are significant individual differences in the efficacy of commonly used sedatives, analgesics and muscle relaxants for general anesthesia. Except for non-genetic factors such as gender, age, liver and renal function [17], gene polymorphisms associated with receptors, transporters and metabolizing enzymes play a major role in the disparities of the pharmacodynamics and pharmacokinetics of anesthetics [8]. Genetic factors account for 20–95% of the variation in individual responses to anesthetics [9]. Pharmacogenomics is an available and valuable method to explore the relationship between gene mutations and variability in individual responses to anesthetics. Therapeutic options based on gene polymorphisms could not only improve the outcome of treatments, but also reduce the risk of drug related toxicity and other adverse effects. The purpose of this study is to summarize the mechanisms of genetic polymorphisms in human response differences to commonly used sedatives, analgesics and muscle relaxants, and provide a scientific theoretical basis for the formulation of a rational anesthesia regimen for patients with various genotypes, and ultimately improve the anesthesia quality and avoid the occurrence of potential complications.

Pharmacogenomics of sedatives

Intravenous anesthetics

Propofol, an ultrafast acting intravenous anesthetic agent, is most frequently utilized in induction and maintenance of general anesthesia, as well the sedation for some unpleasant maneuvers of diagnoses and treatments [10,11]. It works mainly by activating gamma-aminobutyric acid (GABA) receptors [12]. Some metabolic enzymes, such as cytochrome P450 2B6 (CYP2B6), cytochrome P450 2C9 (CYP2C9) and UDP-glucuronosyltransferase 1A9 (UGT1A9), are convinced to be involved in the pharmacokinetics of propofol directly or indirectly [1315]. The sensibility of individuals to propofol is complicated, and it is reflected by various drug consumptions and recovery time required [16, 17]. The evidence for a specific association between genetic components and sensibility of propofol in humans remains deficient. Here in this paper CYP2B6 is detailed as an example.

CYP2B6 and propofol

Whether CYP2B6 has an impact on the diverse individual responses to propofol is still controversial. Propofol is initially biotransformed by CYP2B6. Mastrogianni et al. [18] found that the blood propofol distribution at 4 min post-administration in carriers of the T allele was much wider than CYP2B6 c.516G genotypes, which demonstrated CYP2B6 c.516G > T polymorphism was apparently related to the distribution of blood propofol after a single injection. Mourao et al. [19] found that CYP2B6 c.516G > T genetic variant retarded propofol metabolism in 108 healthy adults undergoing total intravenous general anesthesia, resulting in a decrease of approximately 7% in propofol consumption. In addition, a study conducted by Mikstacki et al. [14] further confirmed that polymorphism c.516G > T in the CYP2B6 gene could affect the biotransformation rate of propofol, and CYP2B6 gene might exert a critical role in the optimization of propofol anesthesia. However, some studies were inconsistent with the above results. A pilot study revealed that the mutation of CYP2B6 c.516G > T gene had no significant effect on propofol and were unable to cause obvious individual variability [20]. A prospective study with eighty-three patients enrolled indicated that CYP2B6 gene polymorphism was not an independent factor determining the pharmacokinetics of propofol [21].
The extensive biotransformation of propofol is catalyzed through two pathways, glucuronidation by UDP-glucuronosyltransferase family members and hydroxylation by CYP P450 enzymes [22,23]. 4-Hydroxypropofol, a hydroxylated metabolite of propofol, attributes to one-third of the hypnotic activity of propofol [24]. It is reasonable to propose that the highly polymorphic nature of CYP2B6 makes the expression of corresponding hydroxylation enzymes varied, resulting in diverse concentrations of 4-Hydroxypropofol. Indeed, the ratio between glucuronidation and hydroxylation was described recently as being the subject to the interpatient variation [14]. The elimination of propofol is dependent on both metabolism and distribution, which makes the role of CYP2B6 polymorphisms in the efficacy of propofol controversial [25]. However, observing the present research condition, it can be concluded that the alteration in hydroxylation from CYP2B6 polymorphism is responsible for individual variability of propofol metabolism and efficacy.

Volatile anesthetics

Great advances in inhalation anesthesia have been achieved with the introduction of fluorinated anesthetics. In the group of fluorinated compounds, desflurane and sevoflurane are recently deployed as the most representative agents for inhalation anesthesia in clinical practices [26].

MC1R and desflurane

Desflurane is resistant to biotransformation with an extremely low metabolic rate of less than 0.1%. It can be metabolized into trifluoroacetic acid and inorganic fluorine by CYP2E1 [27]. Besides, according to a study on desflurane requirements in different hair colored female volunteers, the demand for desflurane in red-haired persons was significantly higher than in dark-haired persons, which could be traced to the mutation on the melanocortin-1 receptor (MC1R) genotype. DNA analysis revealed that red haired women carried at least one dysfunctional or weakened MC1R allele, and 80% carried two such alleles [28]. Electroencephalogram (EEG) is a reliable tool for evaluating the depth of sedation induced by volatile or intravenous anesthetics [29]. It was observed that gene polymorphisms of MYD88rs6853, BDNFrs6265 and IL-1βrs1143627 might be associated with the individual variability of EEG in desflurane anesthesia [30].
Human MC1R is a major regulator of melanogenic enzymes. It exists in human melanocytes, glial cells, pituitary tissue, and periaqueductal gray matter [28]. Volatile anesthetics produce the effect of sedation by potentiating inhibitory neurotransmitter receptors and suppressing excitatory receptors in the central nervous system [31]. In fact, the anesthesia potency of inhaled anesthetics, such as desflurane, may be mediated via the spinal cord rather than the higher nervous centers [32,33]. Moreover, although mutations of many MC1R alleles do not affect their function, it has been found that MC1R variants V60L, R142H, R151C, R160W, D294H make MC1R less effective in intracellular cyclic AMP producing after activation [34]. Though these clues are scattered and are from a small number of studies, they shed light on future researches to more clearly explanation of how MC1R gene polymorphisms induce individual variation in desflurane sensitivity.

MDR1 and sevoflurane

Sevoflurane is another widely used volatile anesthetic, and it is well-tolerated for inhalation induction [35]. The sensitivity of sevoflurane varies among different human groups. Ezri et al. [36] suggested that the minimum alveolar concentration (MAC) for sevoflurane differed in various human ethnicities, which appeared to be related to their genetic makeup. Compared with Caucasian Jews, Oriental Jews had less MAC for sevoflurane, and European Jews had even less. What’s more, to ensure an adequate depth of anesthesia and prevent patients from movement during the operation, 24% higher dosage of sevoflurane was required for Jews in Caucasia than in Europe. A study of sevoflurane-remifentanil anesthesia in pediatric tonsillectomy figured out that children bearing multidrug resistant 1 (MDR1) 1236C > T CC genotype received more superior anesthetic effects compared to children with CT + TT genotype, including shorter induction and recovery time, smaller hemodynamic changes at 5 minutes after extubation, better analgesic and sedation effects, as well as less adverse reactions [37].
P-glycoprotein (P-gp), an efflux pump for diverse lipophilic compounds, is encoded by MDR1 [38,39]. Presumedly, gene polymorphisms in MDR1 including 1236C  >  T may alter the expressions of P-gp and make an impact on the absorption of chemicals into cells. Therefore, the pharmacokinetics and the therapeutic efficacy of some medicines can be influenced by MDR1 gene polymorphisms.

GRIN1 and sevoflurane

Chen et al. [40] have systematically screened the molecular sites that might affect the pharmacological action of sevoflurane. They identified that the N-methyl-D-aspartate receptor NR1 subunit gene (GRIN1) polymorphisms (rs28681971 and rs79901440) were intimately related to the time required for unconsciousness induced by sevoflurane. In more details, individuals with the GRIN1 rs28681971 TT genotype took much less time to achieve the target depth of sevoflurane induced anesthesia than those with the TC genotype. And a longer time was spent to obtain the same sedation depth by individuals with the GRIN1 rs79901440 CT genotype than those with the CC genotype.
Volatile anesthetics exert their efficacy by activating GABA receptors and blocking N-methyl-D-aspartate (NMDA) receptors in the central nervous system [41]. GRIN1 is an essential component of NMDA receptor, which has been confirmed by several molecular cloning studies [42,43]. A functional NMDA receptor is the heteromeric complex consisting of two GRIN1 and two GRIN2 subunits. There is a glycine-binding site in GRIN1 subunit [44]. GRIN1 gene mutations (rs28681971 and rs79901440) may impact on the minimum free energy of the molecular’s secondary structure, then disrupt the folding of GRIN1 protein and the function of NMDA receptor [40]. Thus, it is explainable that the action of sevoflurane varies among individuals with mutated GRIN1 genotype. Further cohort study with a larger sample is required to validate these mechanisms.

Pharmacogenomics of opioid analgesics

Adequate analgesia is an indispensable component and the core goal in anesthesia. Dopamine/noradrenalin and endogenous opioids are largely responsible for the activity of the descending pain inhibitory pathways [45]. Analgesics represented by opioids such as fentanyl, sufentanil and remifentanil are used in nearly all surgical procedures [46]. There are obvious individual differences in the efficacy of these opioids, which undoubtedly poses a challenge for effective pain management. In addition to the non-genetic factors, the role of genetic variations in pharmacokinetics of analgesics has attracted extensive attention of researchers in recent years.

Fentanyl

Fentanyl is the first ever synthesized potent lipid-soluble opioid [47]. It has gained popularity for its versatility in numerous acute and chronic pain management, as well as in the induction of general anesthesia and postoperative analgesia. Previous studies have pointed out that metabolic enzymes and transporters play a vital role in the pharmacokinetics of fentanyl. However, the research on the efficacy of fentanyl is affected by multiple factors including discrepancies in races, sample sizes, administration methods etc. The impact of gene polymorphisms on fentanyl requirements is still controversial.

CYP3A4 and fentanyl

MDR1/CYP3A4/OPRM1 gene polymorphisms in Chinese women were identified to influence the consumption of fentanyl during caesarean section and the effect of postoperative intravenous analgesia [48]. The analgesic efficacy of fentanyl is also related to CYP3A4 polymorphisms. Moreover, data from Zhang et al. [49] CYP3A4*1G gene polymorphisms reduced the metabolism of fentanyl, and the level of CYP3A4 mRNA was positively correlated with fentanyl metabolism in liver microsomes.
Fentanyl is metabolized by CYP P450 3A4 (CYP3A4) and CYP P450 3A5 (CYP3A5), two major oxidative enzymes in liver [50,51]. CYP3A4*1G gene polymorphism could attenuate CYP3A activity directly, and reduce the postoperative consumption of fentanyl [52]. However, CYP3A4 polymorphisms may also bring no obvious changes to CYP3A4 activity, while the combination of either the variant alleles of CYP3A4 or CYP3A5*3 could decrease CYP3A5 activity [49]. Thus CYP gene polymorphism is a possible mechanism for an impaired fentanyl metabolism and different responses to it.

UGT2B7 and fentanyl

As the predominant isozyme of UDP-glucuronosyltransferases, uridine diphosphate-glucuronyltransferase 2B7 (UGT2B7) polymorphism is involved in the biotransformation of multitudinous endobiotics and xenobiotics [53,54]. Fentanyl metabolism is of no exception. Studies revealed that UGT2B7 rs7439366 C allele could enhance the effect of fentanyl, which might be ascribed to the impact of UGT2B7 on fentanyl metabolism [53,55]. Patients with UGT2B7 rs7439366 CT genotype had higher fentanyl sensitivity compared to those with UGT2B7 rs7439366 TT genotype [56].
In humans, UGT2B7 is a uridine diphosphate glucuronic acid transferase with a major role in the disposition of a wide array of small endogenous and exogenous molecules [54,57]. Norfentanyl is the metabolite of fentanyl and has little pharmacological activity [55]. Norfentanyl is reportedly divided into M1, M2, M3, M4, M5, and M7 based on its chemical structure. A portion of norfentanyl is excreted by glucuronidation through urine and bile. The rest norfentanyl is metabolized by glucuronate conjugation, which is dominated by UGT2B7 [57]. Gene polymorphisms of UGT2B7 may alter the activity and function of the enzyme, and affect the metabolism and pharmacokinetics of fentanyl [58].

ABCB1 and fentanyl

Fentanyl is a possible substrate of P-gp which is encoded by adenosine triphosphate binding cassette subfamily B member 1 (ABCB1) [59]. Accordingly, metabolism of fentanyl may also be altered by genetic polymorphisms in ABCB1. Furthermore, there is a positive correlation of single-nucleotide polymorphisms in ABCB1 with respiratory suppression by intravenous fentanyl administration [60]. Horvat et al. [61] observed that less fentanyl was needed for children with ABCB1 rs1045642AA genotype than with AG and GG genotype, which could be explained by decreased expression and activity of P-gp.
A gene-association study has clarified that the analgesic effect of opioids could be influenced by a number of gene polymorphisms including ABCB1 [62]. P-gp encoded by ABCB1 is an integral membranous protein that pumps substrates out of the intracellular compartment [60]. It has been investigated that interindividual differences in P-gp expression and activity is related to ABCB1 polymorphisms [63,64]. Against this background, we hypothesize that ABCB1 polymorphisms could change the substrate disposition of P-gp (fentanyl) and influence its clinical efficacy.

Sufentanil

Sufentanil, a piperidine derivative, is 6–10 times more potent than fentanyl [65]. Sufentanil is now widely used for the induction and maintenance of general anesthesia. In all likelihood, the metabolic mechanism of sufentanil is similar to that of fentanyl.

CYP3A4 and sufentanil

Individual genetic background is also a valuable element for the sensitivity of sufentanil. The high frequency of CYP3A4∗1G variants in Chinese population has been well documented. By oxidizing human liver microsomes, CYP3A4∗1G is believed to be responsible for the metabolism of 45–60% of prescribed drugs including analgesic agents [6567]. As expected, Zhang et al. [68] proposed that CYP3A4*1G gene polymorphisms resulted in the attenuation of CYP3A activity and the reduction of sufentanil consumption for intraoperative pain management in general anesthesia. The correlation between CYP3A4 gene polymorphisms and the consumption of sufentanil has been further proved by Lv et al. [69]. Polymorphisms of CYP3A4∗1G gene caused significant heterogeneity of the postoperative analgesic effect with sufentanil among various ethnic groups [70].
Like fentanyl, sufentanil is mainly metabolized in liver by CYP3A4, the most important isoform of CYP P450 enzymes [71]. Gene mutations in CYP3A4 theoretically impair its activity and result in individual differences in the metabolism and antinociceptive effects of sufentanil.

Remifentanil

Remifentanil is an ultra-short-acting and powerful synthetic opioid. With its less side effect profile, remifentanil is widely applied in clinical work, especially in the maintenance of general anesthesia [72].

5-HTTLPR and remifentanil

In recent years, accumulating researches on pharmacogenomics of remifentanil have been successively conducted. Gene polymorphisms of serotonin transporter gene (a functional 43 bp insertion/deletion polymorphism of serotonin transporter gene (5-HTTLPR), rs25531) has been highlighted for the variation of analgesic response to remifentanil [73]. Individuals with 5-HTTLPR low expression genotype responded with more pain relief to remifentanil than individuals with 5-HTTLPR high expression genotype.
Localized in pre-synaptic neuronal membranes, serotonin transporter (5-HTT) is a key regulator of serotonin metabolism and availability. It terminates synaptic actions by transporting serotonin from the synapse back into the pre-synaptic neuron [74]. 5-HTTLPR is a known polymorphism in 5-HTT promoter region and can influence 5-HTT gene transcription [75]. Although the key role of the rostral ventromedial medulla (RVM) in opioidergic pain regulatory mechanism is well known, a separate serotonergic channel from RVM is also crucial in modulating pain transmission in the dorsal horn of the spinal cord [76]. Several animal studies have demonstrated that serotonin is involved with the clinical analgesia of opioids at the spinal cord level [77,78]. It can be inferred that individual variability in pain responses to opioid may put down to changes in 5-HTT function caused by gene polymorphism 5-HTTLPR.

TMEM8A/SLC9A9 and remifentanil

To comprehensively investigate the genetic factors underlying individual differences in remifentanil demand, a multistage genome-wide association study was carried out in patients who underwent laparoscopic-assisted colectomy. It revealed that rs199670311 of encoding transmembrane protein 8A (TMEM8A) gene and rs4839603 of encoding solute carrier family 9 member A9 (SLC9A9) gene were involved in the sensitivity of remifentanil. Carriers of A and T alleles at rs199670311 and rs4839603 were less sensitive to remifentanil during the operation [79]. To date, there have been no studies on remifentanil sensitivity changes of TMEM8A or SLC9A9 that are caused by gene polymorphisms.

Pharmacogenomics of muscle relaxants

Depolarized muscle relaxants

Succinylcholine has been introduced into anesthesia practice for nearly seventy years, and it remains to be of clinical value in some critical conditions due to its unmatchable property of rapid onset and short duration of action [80]. However, concerns over the safety of succinylcholine still remain today.

BChE and succinylcholine

Butyrylcholinesterase enzyme (BChE) is a hydrolase of succinylcholine. More than sixty BChE gene variations are considered to be responsible for the hydrolysis enzyme dysfunction or instability, which leads to approximately 65% of succinylcholine related apnea [81]. The most frequent mutation in the BChE gene is the Kalow (K) variant [82]. Bretlau et al. [83] found the mean duration of succinylcholine induce muscle relaxation was up to 4 minutes longer in K-variant patients than in wild-type patients. Another study showed that variations in the genetic sequence of BChE, especially BChE * I3E4-14C, BChE * FS126 and BChE * 328D, were closely related to prolonged duration of succinylcholine action [82]. Seven new mutation sites of BChE gene (I373T, G467S, W518R, L184S, V421A, M462I and R577H) were further identified as essential biomarkers for prolonged action of succinylcholine [84].
BChE is a serine hydrolase with the highest concentration in plasma and liver [85]. Earlier reports claimed that genetic variant of BCHE gene impaired the quaternary organization of the tetramer, and decreased the plasma concentration and activity of BChE molecules [86]. Furthermore, it has been found that patients with atypical BChE gene variant are unable to be involved in the normal metabolism of muscle relaxants [87]. Herein, what we may conclude is that BCHE gene polymorphisms appear to be a major factor in various sensitivity to succinylcholine and the occurrence of adverse complications related to it.

Non-depolarized muscle relaxants

The wonderful characteristics of succinylcholine are, however, accompanied by many unpleasant, sometimes serious adverse effects. Studies were carried out to find new fast effective non-depolarization muscle relaxant for rapid sequence induction (RSI) of general anesthesia. Among non-depolarizing muscle relaxants, rocuronium is a good option for achieving RSI [88]. Variations of several key genes, including but not limited to organic anion-transporting polypeptide 1B1 (SLCO1B1), organic anion-transporting polypeptide 1A2 (SLCO1A2), ABCB1 and genetic variants of pregnane X-receptor (NR1I2), have considerable consequences for individual pharmacokenetic differences of rocuronium.

SLCO1B1 / SLCO1A2 and rocuronium

Studies found that gene variations in SLCO1B1 and SLCO1A2 could influence the pharmacodynamics of rocuronium. Clinical duration and recovery time were significantly prolonged in patients with SLCO1B1 rs2306283 AG and GG genotypes than in patients with wild-type homozygous genotypes. Similarly, significant reduction of rocuronium elimination and the extension of were observed in patients with SLCO1A2-189-188InsA genotype [5,6].
SLCO1B1 is a kind of membrane transporter predominantly expressed at the basolateral membrane of hepatocytes. It plays a critical role in the hepatic uptake and clearance of numerous ectogenous and endogenous compounds [89]. A possible mechanism was that gene variant might weaken the transport function of SLCO1B1, which decreased the elimination rate of rocuronium in liver. SLCO1A2 is another important member of the membrane SLC proteins family. It is expressed in a substantial number of organs and determines the drug disposition [90]. The clearance of rocuronium is reported to be mediated by SLCO1A2 dependent hepatocellular uptake and biliary excretion [91]. We speculate the activity of OATP1A2 transporter could be changed due to polymorphisms, which appears to contribute to individual differences in sensitivity to rocuronium.

ABCB1 and rocuronium

The role of ABCB1 gene polymorphisms in the response to rocuronium has been well explored. The duration of rocuronium in ABCB1 rs1128503 CT and CC genotype patients were significantly shorter compared to patients in TT genotype, and the recovery time of rocuronium was significantly shorter in ABCB1 rs1128503 CC genotype patients than in CT and TT genotype patients [5]. Similarly, Qi et al. [4] conducted a study on ABCB1 gene polymorphisms in Chinese people. They found that genetic variants ABCB1 rs12720464 and rs1055302 were the vital factors contributing to the individual variability on muscle relaxation recovery.
Transporter P-gp encoded by ABCB1 has a broad spectrum of substrates including rocuronium, which can be transported by P-gp from hepatocytes to gallbladder [92]. Highly polymorphic ABCB1 gene is associated with structural and functional alterations of P-gp [93]. Moreover, ABCB1 rs1045642 C>T gene mutation could cause P-gp transporter dysfunction and interfere with the absorption and excretion mechanisms of rocuronium [94]. Accordingly, abnormal P-gp is likely to influence the biotransformation of rocuronium.

NR1I2 and rocuronium

Recent findings indicated NR1I2 gene variant might be another major contributor to the variability in rocuronium responses. The clinical duration of rocuronium were extended in NR1I2 rs2472677 and rs6785049 genotypes patients [95]. Existing results of clinical trials are too few, and the association between NR1I2 polymorphisms and individual differences of rocuronium deserves further investigation.
Pregnane X receptor encoded by NR1I2 gene is a member of the nuclear hormone receptors family [96]. It has been well documented that nuclear receptors serve as key transcription regulators to modulate the expression of enzymes and transporters which are involved in the degradation of some drugs [97,98]. Judging this background, the mutation of NR1I2 can change the activities of various transporters (ABCB1, OATP1B1, OATP1A2) and metabolizing enzymes (CYP3A4, CYP2C19, UGT1A1) that are involved in the elimination of rocuronium in vivo.
An individualized anesthesia scheme based on the patient’s unique characteristics is essential to the successful operation performing and the early recovery after surgery. These characteristics include not only the clinical features such as the patient’s age, gender, height, weight, et al., but also the profound genetic features. For instance, malignant hyperthermia induced by muscle relaxants and volatile anesthetics results in a high incidence of mortality. By gene analysis, patients who are susceptible to the disease can be identified. Use of opioids can cause severe nausea and vomiting, or delayed respiratory inhibition. With pharmacogenomics, those patients who are of high risk to opioids induced adverse effects can be detected. Therefore, more effective anesthesia and analgesia regimen can be made and a variety of adverse complications that are detrimental to patients’ health both in the short and long term can be prevented. Over the years, studies on pharmacogenomics of narcotics are relatively few. Nevertheless, the limited findings from these researches are quite helpful and illuminating for individualizing drug administration, improving the efficacy and avoiding serious adverse effects. With the unique genetic background of an individual, a tailored anesthesia scheme could be made, which can maximize beneficial effects, minimize side effects, and relieve financial burden. We believe gene information is expected to become a part of the electronic medical record in the near future, which will be especially useful for personalized anesthesia practicing.

NOTES

Funding

None.

Conflicts of Interest

No potential conflict of interest relevant to this article was reported.

Data Availability

The datasets generated during and/or analyzed during the current study are available in the PubMed.

Author Contributions

Yan Sun (Conceptualization; Validation; Writing – original draft; Writing – review & editing)

Hongyu Zhu (Validation; Writing – original draft)

Elham Esmaeili (Conceptualization; Validation)

Xue Tang (Validation)

Zhilin Wu (Validation; Writing – review & editing)

References

1. Mencke T, Schreiber JU, Knoll H, Werth M, Grundmann U, Rensing H. Influence of gender on the intubation conditions with rocuronium. Anaesthesist 2005; 54: 884-8.
crossref pmid pdf
2. van Miert MM, Eastwood NB, Boyd AH, Parker CJ, Hunter JM. The pharmacokinetics and pharmacodynamics of rocuronium in patients with hepatic cirrhosis. Br J Clin Pharmacol 1997; 44: 139-44.
crossref pmid pmc pdf
3. Staals LM, Snoeck MM, Driessen JJ, van Hamersvelt HW, Flockton EA, van den Heuvel MW, et al. Reduced clearance of rocuronium and sugammadex in patients with severe to end-stage renal failure: a pharmacokinetic study. Br J Anaesth 2010; 104: 31-9.
crossref pmid
4. Qi TL, Zhou YH, Yuan JJ, Li ZS, Wang ZY, Chang YZ, et al. Effect of ABCB1 rs12720464 and rs1055302 polymorphisms in Chinese patients on the time course of action of rocuronium administered as a single dose. Int J Clin Pharmacol Ther 2016; 54: 462-70.
crossref pmid
5. Mei Y, Wang SY, Li Y, Yi SQ, Wang CY, Yang M, et al. Role of SLCO1B1, ABCB1, and CHRNA1 gene polymorphisms on the efficacy of rocuronium in Chinese patients. J Clin Pharmacol 2015; 55: 261-8.
crossref pmid
6. Costa AC, Coelho EB, Lanchote VL, Correia BV, Abumansur JT, Lauretti GR, et al. The SLCO1A2 -189_-188InsA polymorphism reduces clearance of rocuronium in patients submitted to elective surgeries. Eur J Clin Pharmacol 2017; 73: 957-63.
crossref pmid pdf
7. Cubells JF, Blanchard JS, Makman MH. The effects of in vivo inactivation of GABA-transaminase and glutamic acid decarboxylase on levels of GABA in the rat retina. Brain Res 1987; 419: 208-15.
crossref
8. Ahmed S, Zhou Z, Zhou J, Chen SQ. Pharmacogenomics of drug metabolizing enzymes and transporters: relevance to precision medicine. Genomics Proteomics Bioinformatics 2016; 14: 298-313.
crossref pmid pmc
9. Kalow W, Tang BK, Endrenyi L. Hypothesis: comparisons of inter- and intra-individual variations can substitute for twin studies in drug research. Pharmacogenetics 1998; 8: 283-9.
crossref pmid
10. Dinis-Oliveira RJ. Metabolic profiles of propofol and fospropofol: clinical and forensic interpretative aspects. Biomed Res Int 2018; 2018: 6852857.
crossref pmid pmc pdf
11. Walsh CT. Propofol: milk of amnesia. Cell 2018; 175: 10-3.
crossref pmid
12. Lundstrom S, Twycross R, Mihalyo M, Wilcock A. Propofol. J Pain Symptom Manage 2010; 40: 466-70.
crossref pmid
13. Zhong Q, Chen X, Zhao Y, Liu R, Yao S. Association of polymorphisms in pharmacogenetic candidate genes with propofol susceptibility. Sci Rep 2017; 7: 3343.
crossref pmid pmc pdf
14. Mikstacki A, Zakerska-Banaszak O, Skrzypczak-Zielinska M, Tamowicz B, Prendecki M, Dorszewska J, et al. The effect of UGT1A9, CYP2B6 and CYP2C9 genes polymorphism on individual differences in propofol pharmacokinetics among Polish patients undergoing general anaesthesia. J Appl Genet 2017; 58: 213-20.
crossref pmid pmc pdf
15. Pavlovic D, Budic I, Jevtovic Stoimenov T, Stokanovic D, Marjanovic V, Stevic M, et al. The effect of UGT1A9, CYP2B6 and CYP2C9 genes polymorphism on propofol pharmacokinetics in children. Pharmgenomics Pers Med 2020; 13: 13-27.
crossref pmid pmc
16. Iohom G, Ni Chonghaile M, O'Brien JK, Cunningham AJ, Fitzgerald DF, Shields DC. An investigation of potential genetic determinants of propofol requirements and recovery from anaesthesia. Eur J Anaesthesiol 2007; 24: 912-9.
crossref pmid
17. Ypsilantis P, Politou M, Mikroulis D, Lambropoulou M, Bougioukas I, Theodoridis G, et al. Attenuation of propofol tolerance conferred by remifentanil co-administration does not reduce propofol toxicity in rabbits under prolonged mechanical ventilation. J Surg Res 2011; 168: 253-61.
crossref pmid
18. Mastrogianni O, Gbandi E, Orphanidis A, Raikos N, Goutziomitrou E, Kolibianakis EM, et al. Association of the CYP2B6 c.516G>T polymorphism with high blood propofol concentrations in women from northern Greece. Drug Metab Pharmacokinet 2014; 29: 215-8.
crossref pmid
19. Mourao AL, de Abreu FG, Fiegenbaum M. Impact of the Cytochrome P450 2B6 (CYP2B6) Gene Polymorphism c.516G>T (rs3745274) on propofol dose variability. Eur J Drug Metab Pharmacokinet 2016; 41: 511-5.
crossref pmid pdf
20. Loryan I, Lindqvist M, Johansson I, Hiratsuka M, van der Heiden I, van Schaik RH, et al. Influence of sex on propofol metabolism, a pilot study: implications for propofol anesthesia. Eur J Clin Pharmacol 2012; 68: 397-406.
crossref pmid pdf
21. Kanaya A, Sato T, Fuse N, Yamaguchi H, Mano N, Yamauchi M. Impact of clinical factors and UGT1A9 and CYP2B6 genotype on inter-individual differences in propofol pharmacokinetics. J Anesth 2018; 32: 236-43.
crossref pmid pdf
22. Girard H, Court MH, Bernard O, Fortier LC, Villeneuve L, Hao Q, et al. Identification of common polymorphisms in the promoter of the UGT1A9 gene: evidence that UGT1A9 protein and activity levels are strongly genetically controlled in the liver. Pharmacogenetics 2004; 14: 501-15.
crossref pmid
23. Court MH, Duan SX, Hesse LM, Venkatakrishnan K, Greenblatt DJ. Cytochrome P-450 2B6 is responsible for interindividual variability of propofol hydroxylation by human liver microsomes. Anesthesiology 2001; 94: 110-9.
crossref pmid pdf
24. Oda Y, Hamaoka N, Hiroi T, Imaoka S, Hase I, Tanaka K, et al. Involvement of human liver cytochrome P4502B6 in the metabolism of propofol. Br J Clin Pharmacol 2001; 51: 281-5.
crossref pmid pmc pdf
25. Kanto J, Gepts E. Pharmacokinetic implications for the clinical use of propofol. Clin Pharmacokinet 1989; 17: 308-26.
crossref pmid
26. Markuliak RČLHM. General inhalational anesthetics - pharmacodynamics, pharmacokinetics and chiral properties. Ceska Slov Farm 2021; 70: 7-17.
pmid
27. Restrepo JG, Garcia-Martin E, Martinez C, Agundez JA. Polymorphic drug metabolism in anaesthesia. Curr Drug Metab 2009; 10: 236-46.
crossref pmid
28. Liem EB, Lin CM, Suleman MI, Doufas AG, Gregg RG, Veauthier JM, et al. Anesthetic requirement is increased in redheads. Anesthesiology 2004; 101: 279-83.
crossref pmid pmc pdf
29. Jameson LC, Sloan TB. Using EEG to monitor anesthesia drug effects during surgery. J Clin Monit Comput 2006; 20: 445-72.
crossref pmid pdf
30. Mulholland CV, Somogyi AA, Barratt DT, Coller JK, Hutchinson MR, Jacobson GM, et al. Association of innate immune single-nucleotide polymorphisms with the electroencephalogram during desflurane general anaesthesia. J Mol Neurosci 2014; 52: 497-506.
crossref pmid pdf
31. Abdel-Malek ZA. Melanocortin receptors: their functions and regulation by physiological agonists and antagonists. Cell Mol Life Sci 2001; 58: 434-41.
crossref pmid pdf
32. Rampil IJ, Mason P, Singh H. Anesthetic potency (MAC) is independent of forebrain structures in the rat. Anesthesiology 1993; 78: 707-12.
crossref pmid pdf
33. Grasshoff C, Rudolph U, Antkowiak B. Molecular and systemic mechanisms of general anaesthesia: the 'multi-site and multiple mechanisms' concept. Curr Opin Anaesthesiol 2005; 18: 386-91.
crossref pmid
34. Schaffer JV, Bolognia JL. The melanocortin-1 receptor: red hair and beyond. Arch Dermatol 2001; 137: 1477-85.
crossref pmid
35. Palanca BJ, Avidan MS, Mashour GA. Human neural correlates of sevoflurane-induced unconsciousness. Br J Anaesth 2017; 119: 573-82.
crossref pmid pmc
36. Ezri T, Sessler D, Weisenberg M, Muzikant G, Protianov M, Mascha E, et al. Association of ethnicity with the minimum alveolar concentration of sevoflurane. Anesthesiology 2007; 107: 9-14.
crossref pmid pdf
37. Shi NJ, Zhang WX, Zhang N, Zhong LN, Wang LP. Correlation of MDR1 gene polymorphisms with anesthetic effect of sevoflurane-remifentanil following pediatric tonsillectomy. Medicine (Baltimore) 2017; 96: e7002.
crossref pmid pmc
38. Kesimci E, Engin AB, Kanbak O, Karahalil B. Association between ABCB1 gene polymorphisms and fentanyl's adverse effects in Turkish patients undergoing spinal anesthesia. Gene 2012; 493: 273-7.
crossref pmid
39. Byon HJ, Park KS, Park YH, Kim JT, Jung CW, Kim HS. The influence of DNA polymorphism of multidrug resistant 1 (MDR1) on the effect of midazolam pretreatment in children. Korean J Anesthesiol 2012; 62: 332-6.
crossref pmid pmc
40. Chen MH, Ouyang W, Xia YH, Zeng YJ, Wang SY, Duan KM, et al. Association between well-characterized gene polymorphisms and the hypnosis response caused by sevoflurane-induced anaesthesia. J Clin Pharm Ther 2020; 45: 1442-51.
crossref pmid pdf
41. Vutskits L, Xie Z. Lasting impact of general anaesthesia on the brain: mechanisms and relevance. Nat Rev Neurosci 2016; 17: 705-17.
crossref pmid pdf
42. Meguro H, Mori H, Araki K, Kushiya E, Kutsuwada T, Yamazaki M, et al. Functional characterization of a heteromeric NMDA receptor channel expressed from cloned cDNAs. Nature 1992; 357: 70-4.
crossref pmid pdf
43. Chatterton JE, Awobuluyi M, Premkumar LS, Takahashi H, Talantova M, Shin Y, et al. Excitatory glycine receptors containing the NR3 family of NMDA receptor subunits. Nature 2002; 415: 793-8.
crossref pmid pdf
44. Petrenko AB, Yamakura T, Sakimura K, Baba H. Defining the role of NMDA receptors in anesthesia: are we there yet? Eur J Pharmacol 2014; 723: 29-37.
crossref pmid
45. Edwards RR. Genetic predictors of acute and chronic pain. Curr Rheumatol Rep 2006; 8: 411-7.
crossref pmid pdf
46. Scholz J, Steinfath M, Schulz M. Clinical pharmacokinetics of alfentanil, fentanyl and sufentanil. An update. Clin Pharmacokinet 1996; 31: 275-92.
crossref pmid
47. Stanley TH. Fentanyl. J Pain Symptom Manage 2005; 29(5 Suppl): S67-71.
crossref
48. Zhang J, Zhang L, Zhao X, Shen S, Luo X, Zhang Y. Association between MDR1/CYP3A4/OPRM1 gene polymorphisms and the post-caesarean fentanyl analgesic effect on Chinese women. Gene 2018; 661: 78-84.
crossref pmid
49. Zhang W, Yuan JJ, Kan QC, Zhang LR, Chang YZ, Wang ZY, et al. Influence of CYP3A5*3 polymorphism and interaction between CYP3A5*3 and CYP3A4*1G polymorphisms on post-operative fentanyl analgesia in Chinese patients undergoing gynaecological surgery. Eur J Anaesthesiol 2011; 28: 245-50.
crossref pmid
50. Guitton J, Buronfosse T, Desage M, Lepape A, Brazier JL, Beaune P. Possible involvement of multiple cytochrome P450S in fentanyl and sufentanil metabolism as opposed to alfentanil. Biochem Pharmacol 1997; 53: 1613-9.
crossref pmid
51. Feierman DE, Melinkov Z, Nanji AA. Induction of CYP3A by ethanol in multiple in vitro and in vivo models. Alcohol Clin Exp Res 2003; 27: 981-8.
crossref pmid
52. Zhang W, Chang YZ, Kan QC, Zhang LR, Li ZS, Lu H, et al. CYP3A4*1G genetic polymorphism influences CYP3A activity and response to fentanyl in Chinese gynecologic patients. Eur J Clin Pharmacol 2010; 66: 61-6.
crossref pmid pdf
53. Ning M, Tao Y, Hu X, Guo L, Ni J, Hu J, et al. Roles of UGT2B7 C802T gene polymorphism on the efficacy of morphine treatment on cancer pain among the Chinese han population. Niger J Clin Pract 2019; 22: 1319-23.
crossref pmid
54. Shen ML, Xiao A, Yin SJ, Wang P, Lin XQ, Yu CB, et al. Associations between UGT2B7 polymorphisms and cancer susceptibility: a meta-analysis. Gene 2019; 706: 115-23.
crossref pmid
55. Muraoka W, Nishizawa D, Fukuda K, Kasai S, Hasegawa J, Wajima K, et al. Association between UGT2B7 gene polymorphisms and fentanyl sensitivity in patients undergoing painful orthognathic surgery. Mol Pain 2016; 12: 1744806916683182.
crossref pmid pmc pdf
56. Yang Z, Yin Q, Li X. Influences of UGT2B7 rs7439366 and rs12233719 polymorphisms on fentanyl sensitivity in chinese gynecologic patients. Med Sci Monit 2020; 26: e924153.
crossref pmid pmc
57. Coffman BL, King CD, Rios GR, Tephly TR. The glucuronidation of opioids, other xenobiotics, and androgens by human UGT2B7Y(268) and UGT2B7H(268). Drug Metab Dispos 1998; 26: 73-7.
pmid
58. Lu Q, Huang YT, Shu Y, Xu P, Xiang DX, Qu Q, et al. Effects of CYP3A5 and UGT2B7 variants on steady-state carbamazepine concentrations in Chinese epileptic patients. Medicine (Baltimore) 2018; 97: e11662.
crossref pmid pmc
59. Yamazaki M, Neway WE, Ohe T, Chen I, Rowe JF, Hochman JH, et al. In vitro substrate identification studies for p-glycoprotein-mediated transport: species difference and predictability of in vivo results. J Pharmacol Exp Ther 2001; 296: 723-35.
pmid
60. Park HJ, Shinn HK, Ryu SH, Lee HS, Park CS, Kang JH. Genetic polymorphisms in the ABCB1 gene and the effects of fentanyl in Koreans. Clin Pharmacol Ther 2007; 81: 539-46.
crossref pmid
61. Horvat CM, Au AK, Conley YP, Kochanek PM, Li L, Poloyac SM, et al. ABCB1 genotype is associated with fentanyl requirements in critically ill children. Pediatr Res 2017; 82: 29-35.
crossref pmid pmc pdf
62. Zwisler ST, Enggaard TP, Noehr-Jensen L, Mikkelsen S, Verstuyft C, Becquemont L, et al. The antinociceptive effect and adverse drug reactions of oxycodone in human experimental pain in relation to genetic variations in the OPRM1 and ABCB1 genes. Fundam Clin Pharmacol 2010; 24: 517-24.
crossref pmid
63. Hsiao P, Sasongko L, Link JM, Mankoff DA, Muzi M, Collier AC, et al. Verapamil P-glycoprotein transport across the rat blood-brain barrier: cyclosporine, a concentration inhibition analysis, and comparison with human data. J Pharmacol Exp Ther 2006; 317: 704-10.
crossref pmid
64. Takeuchi T, Yoshitomi S, Higuchi T, Ikemoto K, Niwa S, Ebihara T, et al. Establishment and characterization of the transformants stably-expressing MDR1 derived from various animal species in LLC-PK1. Pharm Res 2006; 23: 1460-72.
crossref pmid pdf
65. Maciejewski D. Sufentanil in anaesthesiology and intensive therapy. Anaesthesiol Intensive Ther 2012; 44: 35-41.
pmid
66. Yuan JJ, Hou JK, Zhang W, Chang YZ, Li ZS, Wang ZY, et al. CYP3A4 * 1G genetic polymorphism influences metabolism of fentanyl in human liver microsomes in chinese patients. Pharmacology 2015; 96: 55-60.
crossref
67. Dong ZL, Li H, Chen QX, Hu Y, Wu SJ, Tang LY, et al. Effect of CYP3A4*1G on the fentanyl consumption for intravenous patient-controlled analgesia after total abdominal hysterectomy in Chinese Han population. J Clin Pharm Ther 2012; 37: 153-6.
crossref pmid
68. Zhang H, Chen M, Wang X, Yu S. Patients with CYP3A4*1G genetic polymorphism consumed significantly lower amount of sufentanil in general anesthesia during lung resection. Medicine (Baltimore) 2017; 96: e6013.
crossref pmid pmc
69. Lv J, Liu F, Feng N, Sun X, Tang J, Xie L, et al. CYP3A4 gene polymorphism is correlated with individual consumption of sufentanil. Acta Anaesthesiol Scand 2018; 62: 1367-73.
crossref pmid pdf
70. Zhang C, Zheng Q, Pan F, Wang T, Zhao Y, Xiao Z, et al. Significance of CYP3A4∗1G and OPRM1 A118G polymorphisms in postoperative sufentanil analgesia in women of different ethnicities. Comput Math Methods Med 2022; 2022: 9833591.
crossref pmid pmc
71. Tateishi T, Krivoruk Y, Ueng YF, Wood AJ, Guengerich FP, Wood M. Identification of human liver cytochrome P-450 3A4 as the enzyme responsible for fentanyl and sufentanil N-dealkylation. Anesth Analg 1996; 8: 167-72.
crossref pmid
72. Grape S, Kirkham KR, Frauenknecht J, Albrecht E. Intra-operative analgesia with remifentanil vs. dexmedetomidine: a systematic review and meta-analysis with trial sequential analysis. Anaesthesia 2019; 74: 793-800.
crossref pmid pdf
73. Kosek E, Jensen KB, Lonsdorf TB, Schalling M, Ingvar M. Genetic variation in the serotonin transporter gene (5-HTTLPR, rs25531) influences the analgesic response to the short acting opioid Remifentanil in humans. Mol Pain 2009; 5: 37.
crossref pmid pmc pdf
74. Serretti A, Calati R, Mandelli L, De Ronchi D. Serotonin transporter gene variants and behavior: a comprehensive review. Curr Drug Targets 2006; 7: 1659-69.
crossref pmid
75. Lesch KP, Bengel D, Heils A, Sabol SZ, Greenberg BD, Petri S, et al. Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region. Science 1996; 274: 1527-31.
crossref pmid
76. Fields H. State-dependent opioid control of pain. Nat Rev Neurosci 2004; 5: 565-75.
crossref pmid pdf
77. Hain HS, Belknap JK, Mogil JS. Pharmacogenetic evidence for the involvement of 5-hydroxytryptamine (Serotonin)-1B receptors in the mediation of morphine antinociceptive sensitivity. J Pharmacol Exp Ther 1999; 291: 444-9.
pmid
78. Song B, Chen W, Marvizon JC. Inhibition of opioid release in the rat spinal cord by serotonin 5-HT(1A) receptors. Brain Res 2007; 1158: 57-62.
crossref pmid pmc
79. Nishizawa D, Mieda T, Tsujita M, Nakagawa H, Yamaguchi S, Kasai S, et al. Genome-wide scan identifies candidate loci related to remifentanil requirements during laparoscopic-assisted colectomy. Pharmacogenomics 2018; 19: 113-27.
crossref pmid
80. Sparr HJ, Jöhr M. Succinylcholin-update [Succinylcholine--update]. Anaesthesist 2002; 51: 565-75.
crossref pmid pdf
81. Soliday FK, Conley YP, Henker R. Pseudocholinesterase deficiency: a comprehensive review of genetic, acquired, and drug influences. AANA J 2010; 78: 313-20.
pmid
82. Levano S, Ginz H, Siegemund M, Filipovic M, Voronkov E, Urwyler A, et al. Genotyping the butyrylcholinesterase in patients with prolonged neuromuscular block after succinylcholine. Anesthesiology 2005; 102: 531-5.
crossref pmid pdf
83. Bretlau C, Sørensen MK, Vedersoe AL, Rasmussen LS, Gätke MR. Response to succinylcholine in patients carrying the K-variant of the butyrylcholinesterase gene. Anesth Analg 2013 Mar; 116(3): 596-601.
crossref pmid
84. Wichmann S, Faerk G, Bundgaard JR, Gatke MR. Patients with prolonged effect of succinylcholine or mivacurium had novel mutations in the butyrylcholinesterase gene. Pharmacogenet Genomics 2016; 26: 351-6.
crossref pmid
85. Lockridge O. Review of human butyrylcholinesterase structure, function, genetic variants, history of use in the clinic, and potential therapeutic uses. Pharmacol Ther 2015; 148: 34-46.
crossref pmid
86. Podoly E, Shalev DE, Shenhar-Tsarfaty S, Bennett ER, Ben Assayag E, Wilgus H, et al. The butyrylcholinesterase K variant confers structurally derived risks for Alzheimer pathology. J Biol Chem 2009; 284: 17170-9.
crossref pmid pmc
87. McGuire MC, Nogueira CP, Bartels CF, Lightstone H, Hajra A, Van der Spek AF, et al. Identification of the structural mutation responsible for the dibucaine-resistant (atypical) variant form of human serum cholinesterase. Proc Natl Acad Sci U S A 1989; 86: 953-7.
crossref pmid pmc
88. Guihard B, Chollet-Xemard C, Lakhnati P, Vivien B, Broche C, Savary D, et al. Effect of rocuronium vs succinylcholine on endotracheal intubation success rate among patients undergoing out-of-hospital rapid sequence intubation: a randomized clinical trial. JAMA 2019; 322: 2303-12.
crossref pmid pmc
89. Lee HH, Ho RH. Interindividual and interethnic variability in drug disposition: polymorphisms in organic anion transporting polypeptide 1B1 (OATP1B1; SLCO1B1). Br J Clin Pharmacol 2017; 83: 1176-84.
crossref pmid pmc pdf
90. Zhou Y, Yuan J, Li Z, Wang Z, Cheng D, Du Y, et al. Genetic polymorphisms and function of the organic anion-transporting polypeptide 1A2 and its clinical relevance in drug disposition. Pharmacology 2015; 95: 201-8.
crossref pdf
91. Proost JH, Eriksson LI, Mirakhur RK, Roest G, Wierda JM. Urinary, biliary and faecal excretion of rocuronium in humans. Br J Anaesth 2000; 85: 717-23.
crossref pmid
92. Chen Z, Shi T, Zhang L, Zhu P, Deng M, Huang C, et al. Mammalian drug efflux transporters of the ATP binding cassette (ABC) family in multidrug resistance: A review of the past decade. Cancer Lett 2016; 370: 153-64.
crossref pmid
93. Liu X. ABC family transporters. Adv Exp Med Biol 2019; 1141: 13-100.
crossref pmid
94. Wang SY, Duan KM, Li Y, Mei Y, Sheng H, Liu H, et al. Effect of quercetin on P-glycoprotein transport ability in Chinese healthy subjects. Eur J Clin Nutr 2013; 67: 390-4.
crossref pmid pdf
95. Zhu H, Wang Y, Wang Q, Zhao S, Xu F, Hu Z, et al. Polymorphisms contribute to differences in the effect of rocuronium in Chinese patients. Basic Clin Pharmacol Toxicol 2022; 130: 141-50.
crossref pmid pdf
96. Zhang J, Kuehl P, Green ED, Touchman JW, Watkins PB, Daly A, et al. The human pregnane X receptor: genomic structure and identification and functional characterization of natural allelic variants. Pharmacogenetics 2001; 11: 555-72.
crossref pmid
97. Torres-Vergara P, Ho YS, Espinoza F, Nualart F, Escudero C, Penny J. The constitutive androstane receptor and pregnane X receptor in the brain. Br J Pharmacol 2020; 177: 2666-82.
crossref pmid pmc pdf
98. Elliot ER, Neary M, Else L, Khoo S, Moyle G, Carr DF, et al. Genetic influence of ABCG2, UGT1A1 and NR1I2 on dolutegravir plasma pharmacokinetics. J Antimicrob Chemother 2020; 75: 1259-66.
crossref pmid pdf


ABOUT
ARTICLE CATEGORY

Browse all articles >

BROWSE ARTICLES
AUTHOR INFORMATION
Editorial Office
101-3503, Lotte Castle President, 109 Mapo-daero, Mapo-gu, Seoul 04146, Korea
Tel: +82-2-792-5128    Fax: +82-2-792-4089    E-mail: journal@anesthesia.or.kr                

Copyright © 2024 by Korean Society of Anesthesiologists.

Developed in M2PI

Close layer
prev next