Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 89
1.
Orthop Nurs ; 40(3): 189-193, 2021.
Article En | MEDLINE | ID: mdl-34004619

Ketamine is a dissociative anesthetic used increasingly as analgesia for different manifestations of pain, including acute, chronic, cancer and perioperative pain as well as pain in the critically ill patient population. Its distinctive pharmacologic properties may provide benefits to individuals suffering from pain, including increased pain control and reduction in opioid consumption and tolerance. Despite wide variability in proposed dosing and method of administration when used for analgesia, it is important all clinicians be familiar with the pharmacodynamics of ketamine in order to appropriately anticipate its therapeutic and adverse effects.


Analgesia , Anesthetics, Dissociative , Ketamine , Pain Management , Pain, Postoperative/drug therapy , Analgesics/administration & dosage , Analgesics, Opioid/therapeutic use , Anesthetics, Dissociative/administration & dosage , Anesthetics, Dissociative/pharmacokinetics , Cancer Pain/drug therapy , Humans , Ketamine/administration & dosage , Ketamine/pharmacokinetics , Pain Measurement
2.
Br J Anaesth ; 127(1): 23-31, 2021 07.
Article En | MEDLINE | ID: mdl-33896589

BACKGROUND: Ketamine has cardiac excitatory side-effects. Currently, data on the effects of ketamine and metabolite concentrations on cardiac output are scarce. We therefore developed a pharmacodynamic model derived from data from a randomised clinical trial. The current study is part of a larger clinical study evaluating the potential mitigating effect of sodium nitroprusside on the psychedelic effects of ketamine. METHODS: Twenty healthy male subjects received escalating esketamine and racemic ketamine doses in combination with either placebo or sodium nitroprusside on four visits: (i) esketamine and placebo, (ii) esketamine and sodium nitroprusside, (iii) racemic ketamine and placebo, and (iv) racemic ketamine and sodium nitroprusside. During each visit, arterial blood samples were obtained and cardiac output was measured. Nonlinear mixed-effect modelling was used to analyse the cardiac output time-series data. Ketamine metabolites were added to the model in a sequential manner to evaluate the effects of metabolites. RESULTS: A model including an S-ketamine and S-norketamine effect best described the data. Ketamine increased cardiac output, whereas modelling revealed that S-norketamine decreased cardiac output. No significant effects were detected for R-ketamine, metabolites other than S-norketamine, or sodium nitroprusside on cardiac output. CONCLUSIONS: S-Ketamine, but not R-ketamine, increased cardiac output in a dose-dependent manner. In contrast to S-ketamine, its metabolite S-norketamine reduced cardiac excitation in a dose-dependent manner. CLINICAL TRIAL REGISTRATION: Dutch Cochrane Center 5359.


Anesthetics, Dissociative/chemistry , Anesthetics, Dissociative/pharmacokinetics , Cardiac Output/drug effects , Ketamine/chemistry , Ketamine/pharmacokinetics , Adult , Cardiac Output/physiology , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Healthy Volunteers , Humans , Male , Stereoisomerism , Young Adult
3.
Anesthesiology ; 133(6): 1192-1213, 2020 12 01.
Article En | MEDLINE | ID: mdl-32997732

BACKGROUND: Several models describing the pharmacokinetics of ketamine are published with differences in model structure and complexity. A systematic review of the literature was performed, as well as a meta-analysis of pharmacokinetic data and construction of a pharmacokinetic model from raw data sets to qualitatively and quantitatively evaluate existing ketamine pharmacokinetic models and construct a general ketamine pharmacokinetic model. METHODS: Extracted pharmacokinetic parameters from the literature (volume of distribution and clearance) were standardized to allow comparison among studies. A meta-analysis was performed on studies that performed a mixed-effect analysis to calculate weighted mean parameter values and a meta-regression analysis to determine the influence of covariates on parameter values. A pharmacokinetic population model derived from a subset of raw data sets was constructed and compared with the meta-analytical analysis. RESULTS: The meta-analysis was performed on 18 studies (11 conducted in healthy adults, 3 in adult patients, and 5 in pediatric patients). Weighted mean volume of distribution was 252 l/70 kg (95% CI, 200 to 304 l/70 kg). Weighted mean clearance was 79 l/h (at 70 kg; 95% CI, 69 to 90 l/h at 70 kg). No effect of covariates was observed; simulations showed that models based on venous sampling showed substantially higher context-sensitive half-times than those based on arterial sampling. The pharmacokinetic model created from 14 raw data sets consisted of one central arterial compartment with two peripheral compartments linked to two venous delay compartments. Simulations showed that the output of the raw data pharmacokinetic analysis and the meta-analysis were comparable. CONCLUSIONS: A meta-analytical analysis of ketamine pharmacokinetics was successfully completed despite large heterogeneity in study characteristics. Differences in output of the meta-analytical approach and a combined analysis of 14 raw data sets were small, indicative that the meta-analytical approach gives a clinically applicable approximation of ketamine population parameter estimates and may be used when no raw data sets are available.


Anesthetics, Dissociative/pharmacokinetics , Ketamine/pharmacokinetics , Adult , Child , Humans
4.
Br J Anaesth ; 125(5): 750-761, 2020 11.
Article En | MEDLINE | ID: mdl-32838982

BACKGROUND: Recent studies show activity of ketamine metabolites, such as hydroxynorketamine, in producing rapid relief of depression-related symptoms and analgesia. To improve our understanding of the pharmacokinetics of ketamine and metabolites norketamine, dehydronorketamine, and hydroxynorketamine, we developed a population pharmacokinetic model of ketamine and metabolites after i.v. administration of racemic ketamine and the S-isomer (esketamine). Pharmacokinetic data were derived from an RCT on the efficacy of sodium nitroprusside (SNP) in reducing the psychotomimetic side-effects of ketamine in human volunteers. METHODS: Three increasing i.v. doses of esketamine and racemic ketamine were administered to 20 healthy volunteers, and arterial plasma samples were obtained for measurement of ketamine and metabolites. Subjects were randomised to receive esketamine/SNP, esketamine/placebo, racemic ketamine/SNP, and racemic ketamine/placebo on four separate occasions. The time-plasma concentration data of ketamine and metabolites were analysed using a population compartmental model approach. RESULTS: The pharmacokinetics of ketamine and metabolites were adequately described by a seven-compartment model with two ketamine, norketamine, and hydroxynorketamine compartments and one dehydronorketamine compartment with metabolic compartments in-between ketamine and norketamine, and norketamine and dehydronorketamine main compartments. Significant differences were found between S- and R-ketamine enantiomer pharmacokinetics, with up to 50% lower clearances for the R-enantiomers, irrespective of formulation. Whilst SNP had a significant effect on ketamine clearances, simulations showed only minor effects of SNP on total ketamine pharmacokinetics. CONCLUSIONS: The model is of adequate quality for use in future pharmacokinetic and pharmacodynamic studies into the efficacy and side-effects of ketamine and metabolites. CLINICAL TRIAL REGISTRATION: Dutch Cochrane Center 5359.


Anesthetics, Dissociative/pharmacokinetics , Ketamine/pharmacokinetics , Adult , Anesthetics, Dissociative/administration & dosage , Biotransformation , Computer Simulation , Cross-Over Studies , Double-Blind Method , Drug Compounding , Female , Humans , Injections, Intravenous , Ketamine/administration & dosage , Ketamine/analogs & derivatives , Ketamine/blood , Ketamine/chemistry , Male , Models, Theoretical , Nitroprusside/therapeutic use , Postoperative Complications/prevention & control , Postoperative Complications/psychology , Stereoisomerism , Young Adult
5.
Behav Brain Res ; 390: 112631, 2020 07 15.
Article En | MEDLINE | ID: mdl-32437885

Ketamine induces safe and effective anesthesia and displays unusual cataleptic properties that gave rise to the term dissociative anesthesia. Since 1970, clinicians only utilized the drug as an anesthetic or analgesic for decades, but ketamine was found to have rapid acting antidepressant effects in 1990s. Accumulated evidence exhibits NMDAR antagonism may not be the only mechanism of ketamine. The contributions of AMPA receptor, mTor signal pathway, monoaminergic system, sigma-1 receptor, cholinergic, opioid and cannabinoid systems, as well as voltage-gated calcium channels and hyperpolarization cyclic nucleotide gated channels are discussed for the antidepressant effects. Also the effects of ketamine's enantiomers and metabolites are reviewed. Furthermore ketamine's anesthetic and analgesic mechanisms are briefly revisited. Overall, pharmacology of ketamine, its enantiomers and metabolites is very unique. Insight into multiple mechanisms of action will provide further development and desirable clinical effects of ketamine.


Anesthetics, Dissociative/pharmacology , Antidepressive Agents/pharmacology , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Ketamine/pharmacology , Receptors, Neurotransmitter/drug effects , Anesthetics, Dissociative/administration & dosage , Anesthetics, Dissociative/pharmacokinetics , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/pharmacokinetics , Humans , Ketamine/administration & dosage , Ketamine/pharmacokinetics
6.
J Clin Lab Anal ; 34(4): e23115, 2020 Apr.
Article En | MEDLINE | ID: mdl-31733006

BACKGROUND: We conducted a randomized clinical trial to determine whether adjunctive lidocaine diminishes the incidence of adverse effects in pediatric patients sedated with ketamine. METHODS: This case-control study involved 586 consecutive pediatric patients necessitating anesthesia. Then systolic blood pressure, heart rate, respiratory rate, and blood oxygen saturation were observed. Alanine aminotransferase (ALT), aspartate aminotransferase (AST), urea nitrogen (BUN), and creatinine (Cr) levels were tested. General dose of ketamine, the time of onset and duration of anesthesia and postoperative recovery, anesthesia effect, and adverse reaction were subsequently compared. High-performance liquid chromatography was employed to detect ketamine concentration at different time points after administration, and the postoperative cognition function was further evaluated. RESULTS: Intra- and post-operation, the rising degree of ALT, AST, BUN, and Cr in patients treated with ketamine was higher than those in patients treated with the ketamine-lidocaine complex. General dose of ketamine, the time of onset and duration of anesthesia, postoperative recovery time, and the incidence rate of adverse reaction in patients treated with ketamine-lidocaine complex were lower, but the concentration of ketamine was higher compared to the patients treated with ketamine. In patients treated with the ketamine-lidocaine complex, elimination half-life of ketamine was prolonged, the area under curve was increased, and the plasma clearance rate was decreased relative to those with ketamine alone. CONCLUSIONS: Ketamine combined with lidocaine may be beneficial in shortening the onset of anesthesia, promoting postoperative awake, prolonging elimination half-life, increasing area under curve, and decreasing plasma clearance rate and incidence of adverse reactions.


Anesthesia/methods , Ketamine/administration & dosage , Ketamine/adverse effects , Lidocaine/administration & dosage , Anesthesia/adverse effects , Anesthetics, Dissociative/administration & dosage , Anesthetics, Dissociative/adverse effects , Anesthetics, Dissociative/blood , Anesthetics, Dissociative/pharmacokinetics , Anesthetics, Local/administration & dosage , Anesthetics, Local/adverse effects , Appendectomy , Child , Child, Preschool , Cleft Lip/surgery , Cleft Palate/surgery , Drug Monitoring , Drug Therapy, Combination , Female , Half-Life , Herniorrhaphy , Humans , Injections, Intravenous , Ketamine/blood , Ketamine/pharmacokinetics , Lidocaine/adverse effects , Male , Operative Time , Postoperative Period , Vital Signs
7.
Vet Anaesth Analg ; 46(3): 335-343, 2019 May.
Article En | MEDLINE | ID: mdl-30956016

OBJECTIVE: To investigate the influence of ketamine on plasma cortisol concentration (PCC) in calves. STUDY DESIGN: Prospective, randomized experimental study. ANIMALS: A total of 41 healthy, predominantly cross-bred calves, aged 3-4 months. METHODS: Calves were premedicated with intramuscular xylazine (0.2 mg kg-1) and randomly divided into four groups. The control group (CONT) received saline (after 10, 20 and 30 minutes), whereas groups K1, K2 and K3 were injected intravenously once, twice or thrice, respectively, with 4 mg kg-1 of ketamine at 10 minute intervals. Blood samples were collected at fixed time points and analysed to determine the PCC; furthermore, the plasma concentrations of ketamine and norketamine were assessed after a single ketamine administration in group K1. The pharmacokinetic parameters of ketamine and norketamine were calculated as plasma concentrations versus time. RESULTS: All groups showed significant (p < 0.0001) increases in PCC compared with the baseline value; however, for the first 60 minutes, PCC was significantly higher in the ketamine-treated groups (time × dose effect; K1: p < 0.0001; K2: p = 0.0008; K3: p = 0.0135) than in the CONT group. The group receiving triple ketamine administration exhibited the greatest increase in PCC compared with the baseline level (121.17 ± 33.25 nmol L-1), whereas in the CONT group, the increase in PCC was smaller than the baseline cortisol level (82.67 ± 36.86 nmol L-1). The plasma concentration of ketamine decreased with a half-life of approximately 12 minutes, which was longer than the dose interval. The increase in PCC after triplicate administration might, therefore, have resulted from ketamine/norketamine accumulation rather than from the total dosage. CONCLUSIONS AND CLINICAL RELEVANCE: Our results showed that ketamine increases the plasma concentration of cortisol in xylazine-treated calves. Thus, the previous treatment of subjects needs to be considered in studies using plasma/serum cortisol concentrations as an indicator of pain.


Anesthesia/veterinary , Anesthetics, Dissociative/pharmacology , Cattle/blood , Hydrocortisone/blood , Ketamine/pharmacology , Xylazine/pharmacology , Anesthetics, Dissociative/pharmacokinetics , Animals , Dose-Response Relationship, Drug , Female , Ketamine/administration & dosage , Ketamine/analogs & derivatives , Ketamine/pharmacokinetics , Male , Pain Measurement/veterinary , Premedication , Prospective Studies , Random Allocation , Single-Blind Method , Xylazine/administration & dosage
8.
Drug Test Anal ; 11(1): 68-76, 2019 Jan.
Article En | MEDLINE | ID: mdl-30027605

Ketamine is a glutamate N-methyl-D-aspartate receptor antagonist that is a rapid-acting dissociative anesthetic. It has been proposed as an adjuvant treatment along with other drugs (atropine, midazolam, pralidoxime) used in the current standard of care (SOC) for organophosphate and nerve agent exposures. Ketamine is a pharmaceutical agent that is readily available to most clinicians in emergency departments and possesses a broad therapeutic index with well-characterized effects in humans. The objective of this study was to determine the pharmacokinetic profile of ketamine and its active metabolite, norketamine, in F344 rats following single or repeated intramuscular administrations of subanesthetic levels (7.5 mg/kg or 30 mg/kg) of ketamine with or without the SOC. Following administration, plasma and brain tissues were collected and analyzed using a liquid chromatography-mass spectrometry method to quantitate ketamine and norketamine. Following sample analysis, the pharmacokinetics were determined using non-compartmental analysis. The addition of the current SOC had a minimal impact on the pharmacokinetics of ketamine following intramuscular administration and repeated dosing at 7.5 mg/kg every 90 minutes allows for sustained plasma concentrations above 100 ng/mL. The pharmacokinetics of ketamine with and without the SOC in rats supports further investigation of the efficacy of ketamine co-administration with the SOC following nerve agent exposure in animal models.


Anesthetics, Dissociative/administration & dosage , Anesthetics, Dissociative/pharmacokinetics , Intramuscular Absorption/drug effects , Ketamine/administration & dosage , Ketamine/pharmacokinetics , Anesthetics, Dissociative/blood , Animals , Injections, Intramuscular/methods , Intramuscular Absorption/physiology , Ketamine/blood , Male , Rats , Rats, Inbred F344 , Tandem Mass Spectrometry/methods , Tandem Mass Spectrometry/standards
9.
Expert Opin Drug Metab Toxicol ; 14(7): 709-720, 2018 Jul.
Article En | MEDLINE | ID: mdl-29888644

INTRODUCTION: Although used as an anesthetic drug for decades, ketamine appears to have garnered renewed interest due to its potential therapeutic uses in pain therapy, neurology, and psychiatry. Ketamine undergoes extensive oxidative metabolism by cytochrome P450 (CYP) enzymes. Considerable efforts have been expended to elucidate the ketamine-induced regulation of CYP gene expression. The safety profile of chronic ketamine administration is still unclear. Understanding how ketamine regulates CYP gene expression is clinically meaningful. Areas covered: In this article, the authors provide a brief review of clinical applications of ketamine and its metabolism by CYP enzymes. We discuss the effects of ketamine on the regulation of CYP gene expression, exploring aspects of cytoskeletal remodeling, mitochondrial functions, and calcium homeostasis. Expert opinion: Ketamine may inhibit CYP gene expression through inhibiting calcium signaling, decreasing ATP levels, producing excessive reactive oxygen species, and subsequently perturbing cytoskeletal dynamics. Further research is still needed to avoid possible ketamine-drug interactions during long-term use in the clinic.


Anesthetics, Dissociative/administration & dosage , Cytochrome P-450 Enzyme System/genetics , Ketamine/administration & dosage , Adenosine Triphosphate/metabolism , Anesthetics, Dissociative/adverse effects , Anesthetics, Dissociative/pharmacokinetics , Animals , Calcium Signaling/drug effects , Cytoskeleton/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Ketamine/adverse effects , Ketamine/pharmacokinetics , Reactive Oxygen Species/metabolism
10.
Biomed Res Int ; 2018: 6562309, 2018.
Article En | MEDLINE | ID: mdl-29951541

Eighteen Sprague-Dawley rats were randomly divided into three groups: ketamine group, rhynchophylline group, and ketamine combined with rhynchophylline group (n = 6). The rats of two groups received a single intraperitoneal administration of 30 mg/kg ketamine and 30 mg/kg rhynchophylline, respectively, and the third group received combined intraperitoneal administration of 30 mg/kg ketamine and 30 mg/kg rhynchophylline together. After blood sampling at different time points and processing, the concentrations of ketamine and rhynchophylline in rat plasma were determined by the established ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method. Chromatographic separation was achieved using a UPLC BEH C18 column (2.1 mm × 50 mm, 1.7 µm) with carbamazepine as an internal standard (IS). The initial mobile phase consisted of acetonitrile and water (containing 0.1% formic acid) with gradient elution. Multiple reaction monitoring (MRM) modes of m/z 238.1 → 179.1 for ketamine, m/z 385.3 → 159.8 for rhynchophylline, and m/z 237.3 → 194.3 for carbamazepine (IS) were utilized to conduct quantitative analysis. Calibration curve of ketamine and rhynchophylline in rat plasma demonstrated good linearity in the range of 1-1000 ng/mL (r > 0.995), and the lower limit of quantification (LLOQ) was 1 ng/mL. Moreover, the intra- and interday precision relative standard deviation (RSD) of ketamine and rhynchophylline were less than 11% and 14%, respectively. This sensitive, rapid, and selective UPLC-MS/MS method was successfully applied to pharmacokinetic interaction study of ketamine and rhynchophylline after intraperitoneal administration. The results showed that there may be a reciprocal inhibition between ketamine and rhynchophylline.


Anesthetics, Dissociative/pharmacokinetics , Ketamine/pharmacokinetics , Oxindoles/pharmacokinetics , Animals , Chromatography, High Pressure Liquid , Chromatography, Liquid , Drug Interactions , Random Allocation , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Tandem Mass Spectrometry
11.
J Clin Psychiatry ; 79(2)2018.
Article En | MEDLINE | ID: mdl-29659211

A growing body of literature suggests that ketamine, administered in subanesthetic doses, has early-onset antidepressant action in patients with severe and even treatment-refractory depression. Many case reports, open-label studies, and randomized controlled trials (RCTs) suggest that ketamine may have dramatic antisuicidal effects, as well. This article examines the benefits of ketamine in patients with suicidal ideation with particular focus on the findings of recent RCTs and meta-analyses. Important findings are that a single dose of ketamine is associated with antisuicidal benefits that emerge within an hour of administration and persist for up to a week. The benefits are seen in patients with mild as well as clinically significant suicidal ideation. The benefits are observed in midazolam- as well as saline-controlled trials. Effect sizes are medium to large. The improvement in suicidal ideation is only partly explained by improvement in depression severity. It is concluded that there is consistent evidence that a single dose of ketamine has dramatic antisuicidal action that emerges almost immediately after dosing and persists for at least a week. The short- and intermediate-term safety and efficacy of ketamine as a crisis intervention treatment for suicidal patients merit study. Areas that need research are outlined.


Crisis Intervention/methods , Ketamine , Suicidal Ideation , Suicide Prevention , Suicide , Anesthetics, Dissociative/administration & dosage , Anesthetics, Dissociative/pharmacokinetics , Antidepressive Agents/administration & dosage , Antidepressive Agents/pharmacokinetics , Dose-Response Relationship, Drug , Drug Repositioning , Humans , Ketamine/administration & dosage , Ketamine/pharmacokinetics , Suicide/psychology , Treatment Outcome
12.
Basic Clin Pharmacol Toxicol ; 122(5): 481-488, 2018 May.
Article En | MEDLINE | ID: mdl-29171155

Ketamine and its primary metabolite norketamine attenuate morphine tolerance by antagonising N-methyl-d-aspartate (NMDA) receptors. Ketamine is extensively metabolized to several other metabolites. The major secondary metabolite (2S,6S;2R,6R)-hydroxynorketamine (6-hydroxynorketamine) is not an NMDA antagonist. However, it may modulate nociception through negative allosteric modulation of α7 nicotinic acetylcholine receptors. We studied whether 6-hydroxynorketamine could affect nociception or the effects of morphine in acute or chronic administration settings. Male Sprague Dawley rats received subcutaneous 6-hydroxynorketamine or ketamine alone or in combination with morphine, as a cotreatment during induction of morphine tolerance, and after the development of tolerance induced by subcutaneous minipumps administering 9.6 mg morphine daily. Tail flick, hot plate, paw pressure and rotarod tests were used. Brain and serum drug concentrations were quantified with high-performance liquid chromatography-tandem mass spectrometry. Ketamine (10 mg/kg), but not 6-hydroxynorketamine (10 and 30 mg/kg), enhanced antinociception and decreased rotarod performance following acute administration either alone or combined with morphine. Ketamine efficiently attenuated morphine tolerance. Acutely administered 6-hydroxynorketamine increased the brain concentration of morphine (by 60%), and brain and serum concentrations of 6-hydroxynorketamine were doubled by morphine pre-treatment. This pharmacokinetic interaction did not, however, lead to altered morphine tolerance. Co-administration of 6-hydroxynorketamine 20 mg/kg twice daily did not influence development of morphine tolerance. Even though morphine and 6-hydroxynorketamine brain concentrations were increased after co-administration, the pharmacokinetic interaction had no effect on acute morphine nociception or tolerance. These results indicate that 6-hydroxynorketamine does not have antinociceptive properties or attenuate opioid tolerance in a similar way as ketamine.


Analgesics, Opioid/pharmacology , Anesthetics, Dissociative/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Drug Tolerance , Ketamine/analogs & derivatives , Morphine/pharmacology , Nociceptive Pain/prevention & control , Analgesics, Opioid/blood , Analgesics, Opioid/pharmacokinetics , Anesthetics, Dissociative/blood , Anesthetics, Dissociative/pharmacokinetics , Animals , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Drug Interactions , Ketamine/blood , Ketamine/pharmacokinetics , Ketamine/pharmacology , Male , Morphine/blood , Morphine/pharmacokinetics , Motor Activity/drug effects , Nociception/drug effects , Nociceptive Pain/blood , Nociceptive Pain/physiopathology , Nociceptive Pain/psychology , Pain Threshold/drug effects , Rats, Sprague-Dawley
13.
Anesthesiology ; 125(6): 1103-1112, 2016 12.
Article En | MEDLINE | ID: mdl-27763887

BACKGROUND: At therapeutic concentrations, cytochrome P4502B6 (CYP2B6) is the major P450 isoform catalyzing hepatic ketamine N-demethylation to norketamine in vitro. The CYP2B6 gene is highly polymorphic. The most common variant allele, CYP2B6*6, is associated with diminished hepatic CYP2B6 expression and catalytic activity compared with wild-type CYP2B6*1/*1. CYP2B6.6, the protein encoded by the CYP2B6*6 allele, and liver microsomes from CYP2B6*6 carriers had diminished ketamine metabolism in vitro. This investigation tested whether humans with the CYP2B6*6 allele would have decreased clinical ketamine metabolism and clearance. METHODS: Thirty volunteers with CYP2B6*1/*1, *1/*6, or *6/*6 genotypes (n = 10 each) received a subsedating dose of oral ketamine. Plasma and urine concentrations of ketamine and the major CYP2B6-dependent metabolites were determined by mass spectrometry. Subjects' self-assessment of ketamine effects were also recorded. The primary outcome was ketamine N-demethylation, measured as the plasma norketamine/ketamine area under the curve ratio. Secondary outcomes included plasma ketamine enantiomer and metabolite area under the plasma concentration-time curve, maximum concentrations, apparent oral clearance, and metabolite formation clearances. RESULTS: There was no significant difference between CYP2B6 genotypes in ketamine metabolism or any of the secondary outcome measures. Subjective self-assessment did reveal some differences in energy and level of awareness among subjects. CONCLUSIONS: These results show that while the CYP2B6*6 polymorphism results in diminished ketamine metabolism in vitro, this allelic variant did not affect single, low-dose ketamine metabolism, clearance, and pharmacokinetics in vivo. While in vitro drug metabolism studies may be informative, clinical investigations in general are needed to validate in vitro observations.


Anesthetics, Dissociative/pharmacokinetics , Cytochrome P-450 CYP2B6/genetics , Ketamine/pharmacokinetics , Polymorphism, Genetic/genetics , Adolescent , Adult , Female , Humans , Male , Middle Aged , Young Adult
14.
J Chromatogr A ; 1467: 436-444, 2016 Oct 07.
Article En | MEDLINE | ID: mdl-27485149

Ketamine is often used for anesthesia in veterinary medicine. One possible comedication is the sedative α2-agonist medetomidine. Advantages of that combination are the compensation of side effects of the two drugs and the anesthetic-sparing effect of medetomidine. In vitro studies showed that medetomidine has an inhibitive effect on the formation of norketamine. Norketamine is the first metabolite of ketamine and is also active. It is followed by others like 6-hydroxynorketamine and 5,6-dehydronorketamine (DHNK). In an in vivo pharmacokinetic study Beagle dogs under sevoflurane anesthesia (mean end-tidal concentration 3.0±0.2%) or following medetomidine sedation (450µg/m2) received 4mg/kg racemic ketamine or 2mg/kg S-ketamine. Blood samples were collected between 0 and 900min after drug injection. 50µL aliquots of plasma were pretreated by liquid-liquid extraction prior to analysis of the reconstituted extracts with a robust enantioselective capillary electrophoresis assay using highly sulfated γ-cyclodextrin as chiral selector and electrokinetic sample injection of the analytes from the extract across a short buffer plug without chiral selector. Levels of S- and R-ketamine, S- and R-norketamine, (2S,6S)- and (2R,6R)-hydroxynorketamine and S- and R-DHNK were determined. Data were analyzed with compartmental pharmacokinetic models which included two compartments for the ketamine and norketamine enantiomers and a single compartment for the DHNK and 6-hydroxynorketamine stereoisomers. Medetomidine showed an effect on the formation and elimination of all metabolites. Stereoselectivities were detected for 6-hydroxynorketamine and DHNK, but not for ketamine and norketamine.


Anesthetics, Dissociative/pharmacokinetics , Anesthetics, Inhalation/pharmacology , Electrophoresis, Capillary , Hypnotics and Sedatives/pharmacology , Ketamine/pharmacokinetics , Medetomidine/pharmacology , Methyl Ethers/pharmacology , Animals , Biotransformation , Dogs , Drug Interactions , Female , Ketamine/analogs & derivatives , Ketamine/chemistry , Male , Sevoflurane , Stereoisomerism , gamma-Cyclodextrins/chemistry
15.
Exp Anim ; 65(4): 383-392, 2016 Nov 01.
Article En | MEDLINE | ID: mdl-27263962

While the cage refinement is a necessary step towards improving the welfare of research rats, increasing the complexity and surface area of the living space of an animal may have physiological impacts that need to be taken into consideration. In this study, ketamine (80 mg/kg) and xylazine (10 mg/kg) caused a short duration anesthesia that was significantly decreased in Sprague-Dawley rats housed in multilevel cages (MLC), compared to rats housed in standard cages (SDC). The withdrawal reflex, the palpebral reflexes and the time-to-sternal all occurred earlier in MLC housed rats, suggesting an effect of housing on the physiology of the rats. In addition, during anesthesia, cardiac frequencies were increased in animals housed in the smaller SDC. Respiratory frequencies, the blood oxygen saturation and rectal temperatures during anesthesia did not vary between conditions during the anesthesia. While xylazine pharmacokinetics were unchanged with caging conditions, the clearance and half-lives of ketamine and its metabolite, norketamine, were altered in the rats housed in MLC. Finally, while no difference was ultimately seen in rat body weights, isolated liver and adrenal gland weights were significantly lighter in rats housed in the MLC. Increasing cage sizes, while having a positive impact on wellbeing in rats, can alter anesthetic drug metabolism and thus modify anesthesia parameters and associated physiological processes.


Anesthetics, Dissociative/pharmacokinetics , Housing, Animal , Hypnotics and Sedatives/pharmacokinetics , Ketamine/pharmacokinetics , Rats/physiology , Xylazine/pharmacokinetics , Anesthetics, Dissociative/pharmacology , Animals , Half-Life , Hypnotics and Sedatives/pharmacology , Ketamine/analogs & derivatives , Ketamine/pharmacology , Male , Rats, Sprague-Dawley , Reflex/drug effects , Specific Pathogen-Free Organisms , Xylazine/pharmacology
16.
Can J Vet Res ; 80(2): 141-5, 2016 Apr.
Article En | MEDLINE | ID: mdl-27127341

This study investigated the effects of ketamine and lidocaine in combination on the minimum alveolar concentration of sevoflurane (MACSEVO) in alpacas. Eight healthy, intact male, adult alpacas were studied on 2 separate occasions. Anesthesia was induced with SEVO, and baseline MAC (MACB) determination began 45 min after induction. After MACB determination, alpacas were randomly given either an intravenous (IV) loading dose (LD) and infusion of saline or a loading dose [ketamine = 0.5 mg/kg body weight (BW); lidocaine = 2 mg/kg BW] and an infusion of ketamine (25 µg/kg BW per minute) in combination with lidocaine (50 µg/kg BW per minute), and MACSEVO was re-determined (MACT). Quality of recovery, time-to-extubation, and time-to-standing, were also evaluated. Mean MACB was 1.88% ± 0.13% and 1.89% ± 0.14% for the saline and ketamine + lidocaine groups, respectively. Ketamine and lidocaine administration decreased (P < 0.05) MACB by 57% and mean MACT was 0.83% ± 0.10%. Saline administration did not change MACB. Time to determine MACB and MACT was not significantly different between the treatments. The quality of recovery, time-to-extubation, and time-to-standing, were not different between groups. The infusion of ketamine combined with lidocaine significantly decreased MACSEVO by 57% and did not adversely affect time-to-standing or quality of recovery.


La présente étude visait à examiner les effets d'une combinaison de kétamine et de lidocaïne sur la concentration alvéolaire minimale de sevoflurane (CAMSEVO) chez des alpagas. Huit alpagas mâles entiers et en santé ont été étudiés en deux occasions distinctes. L'anesthésie a été induite avec du SEVO, et la détermination de la CAM de base (CAMB) débutée 45 min après l'induction. Après détermination de la CAMB, les alpagas ont reçu par voie intraveineuse (IV), sur une base aléatoire, une dose de charge (DC) et une infusion de saline ou une dose de [kétamine = 0,5 mg/kg de poids corporel (PC); lidocaïne = 2 mg/kg PC] et une infusion de kétamine (25 µg/kg PC par minute) en combinaison avec de la lidocaïne (50 µg/kg PC par minute), et la CAMSEVO re-déterminée (CAMT). La qualité de la récupération, le temps pour extuber, et le temps pour se tenir debout ont également été évalués. La CAMB moyenne était de 1,88 % ± 0,13 % et de 1,89 % ± 0,14 % pour les groupes saline et kétamine + lidocaïne, respectivement. L'administration de kétamine et de lidocaïne entraîna une diminution (P < 0,05) de 57 % de CAMB et la CAMT moyenne était de 0,83 % ± 0,10 %. L'administration de saline n'a pas changé la CAMB. Le temps pour déterminer la CAMB et la CAMT n'était pas significativement différent entre les groupes de traitement. La qualité de la récupération, le temps pour extuber, et le temps pour se tenir debout n'étaient pas significativement différents entre les groupes. L'infusion de kétamine combinée à la lidocaïne a diminué significativement la CAMSEVO de 57 % et n'affecta pas négativement le temps pour se tenir debout ou la qualité de la récupération.(Traduit par Docteur Serge Messier).


Anesthesia, Inhalation/veterinary , Camelids, New World , Ketamine/pharmacokinetics , Lidocaine/pharmacokinetics , Methyl Ethers/pharmacokinetics , Pulmonary Alveoli/metabolism , Anesthetics, Dissociative/administration & dosage , Anesthetics, Dissociative/pharmacokinetics , Anesthetics, Dissociative/pharmacology , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/pharmacology , Anesthetics, Local/administration & dosage , Anesthetics, Local/pharmacokinetics , Anesthetics, Local/pharmacology , Animals , Cross-Over Studies , Drug Interactions , Ketamine/administration & dosage , Ketamine/pharmacology , Lidocaine/administration & dosage , Lidocaine/pharmacology , Male , Methyl Ethers/administration & dosage , Methyl Ethers/pharmacology , Sevoflurane
17.
Paediatr Anaesth ; 25(2): 211-6, 2015 Feb.
Article En | MEDLINE | ID: mdl-25212712

BACKGROUND: Intravenous racemic ketamine is commonly administered for procedural sedation, although few pharmacokinetic studies have been conducted among children. Moreover, an optimal sampling schedule has not been derived to enable the conduct of pharmacokinetic studies that minimally inconvenience study participants. METHODS: Concentration-time data were obtained from 57 children who received 1-1.5 mg·kg(-1) of racemic ketamine as an intravenous bolus. A population pharmacokinetic analysis was conducted using nonlinear mixed effects models, and the results were used as inputs to develop a D-optimal sampling schedule. RESULTS: The pharmacokinetics of ketamine were described using a two-compartment model. The volume of distribution in the central and peripheral compartments were 20.5 l∙70 kg(-1) and 220 l∙70 kg(-1), respectively. The intercompartmental clearance and total body clearance were 87.3 and 87.9 l·h(-1) ∙70 kg(-1), respectively. Population parameter variability ranged from 34% to 98%. Initially, blood samples were drawn on 3-6 occasions spanning a range of 14-152 min after dosing. Using these data, we determined that four optimal sampling windows occur at 1-5, 5.5-7.5, 10-20, and 90-180 min after dosing. Monte Carlo simulations indicated that these sampling windows produced precise and unbiased ketamine pharmacokinetic parameter estimates. CONCLUSION: An optimal sampling schedule was developed that allowed assessment of the pharmacokinetic parameters of ketamine among children requiring short-term procedural sedation.


Analgesia/methods , Anesthetics, Dissociative/blood , Anesthetics, Dissociative/pharmacokinetics , Ketamine/blood , Ketamine/pharmacokinetics , Child , Child, Preschool , Computer Simulation , Female , Humans , Injections, Intravenous , Male , Models, Statistical , Monte Carlo Method , Time Factors
18.
Int J Pharm ; 478(1): 223-231, 2015 Jan 15.
Article En | MEDLINE | ID: mdl-25448584

This study aims at developing a population pharmacokinetic model for ketamine in children with cardiac diseases in order to rationalize an effective 2-h anesthetic medication, personalized based on cardiac function and age. Twenty-one children (6 months to 18 years old) were enrolled in this prospective, open label study. Ketamine 2mg/kg IV was administered and blood samples were then collected over 8h for ketamine assay. Pharmacokinetic data analysis using NONMEM, was undertaken. Ketamine pharmacokinetics was adequately described by a two-compartment linear disposition model. Typical population parameters were: total clearance: 60.6 ×(weight/70)(0.75)L/h, intercompartmental clearance: 73.2 ×(weight/70)(0.75)L/h, central distribution volume: 57.3 ×(weight/70)L, and peripheral distribution volume: 152 ×(weight/70)L. Ketamine clearance in children with pre-existing congenital heart disease was comparable to values reported in healthy subjects. Computer simulations indicated that an initial loading dose of ketamine 2mg/kg IV over 1 min followed by a constant rate infusion of 6.3mg/kg/h for 29 min, 4.5mg/kg/h from 30 to 80 min, and 3.9 mg/kg/h from 80 to 120 min achieves and maintains anesthetic plasma level for 2h in children 1 year or older (weight ≥ 10 kg).


Anesthetics, Dissociative/pharmacokinetics , Heart Diseases/metabolism , Ketamine/pharmacokinetics , Adolescent , Anesthetics, Dissociative/blood , Child , Child, Preschool , Female , Heart Diseases/blood , Humans , Infant , Ketamine/blood , Male , Models, Biological
19.
Can J Vet Res ; 78(3): 207-13, 2014 Jul.
Article En | MEDLINE | ID: mdl-24982552

The objective of this study was to determine if prior measurement of the minimum alveolar concentration (MAC) of isoflurane influences the effect of ketamine on the MAC of isoflurane in dogs. Eight mixed-breed dogs were studied on 2 occasions. Anesthesia was induced and maintained using isoflurane. In group 1 the effect of ketamine on isoflurane MAC was determined after initially finding the baseline isoflurane MAC. In group 2, the effect of ketamine on isoflurane MAC was determined without previous measure of the baseline isoflurane MAC. In both groups, MAC was determined again 30 min after stopping the CRI of ketamine. Plasma ketamine concentrations were measured during MAC determinations. In group 1, baseline MAC (mean ± SD: 1.18 ± 0.14%) was decreased by ketamine (0.88 ± 0.14%; P < 0.05). The MAC after stopping ketamine was similar (1.09 ± 0.16%) to baseline MAC and higher than with ketamine (P < 0.05). In group 2, the MAC with ketamine (0.79 ± 0.11%) was also increased after stopping ketamine (1.10 ± 0.17%; P < 0.05). The MAC values with ketamine were different between groups (P < 0.05). Ketamine plasma concentrations were similar between groups during the events of MAC determination. The MAC of isoflurane during the CRI of ketamine yielded different results when methods of same day (group-1) versus separate days (group-2) are used, despite similar plasma ketamine concentrations with both methods. However, because the magnitude of this difference was less than 10%, either method of determining MAC is deemed acceptable for research purposes.


L'objectif de la présente étude était de déterminer si une mesure antérieure de la concentration alvéolaire minimum (MAC) d'isoflurane influence l'effet de la kétamine sur la MAC d'isoflurane chez les chiens. Huit chiens de race croisée ont été examinés à deux occasions. L'anesthésie fut induite et maintenue à l'aide d'isoflurane. Dans le groupe 1, l'effet de la kétamine sur la MAC d'isoflurane fut déterminé après avoir initialement trouvé la MAC de base de l'isoflurane. Dans le groupe 2, l'effet de la kétamine sur la MAC d'isoflurane fut déterminé sans mesure préalable de la MAC de base de l'isoflurane. Dans les deux groupes la MAC fut déterminée de nouveau 30 min après l'arrêt de la CRI de kétamine. Les concentrations de kétamine plasmatiques furent mesurées durant les déterminations de MAC.Dans le groupe 1, la MAC de base (moyenne ± SD : 1,18 ± 0,14 %) fut diminuée par la kétamine (0,88 ± 0,14 %; P < 0,05). La MAC après l'arrêt de la kétamine était similaire (1,09 ± 0,16 %) à la MAC de base et plus élevée qu'avec la kétamine (P < 0,05). Dans le groupe 2, la MAC avec kétamine (0,79 ± 0,11 %) était également augmentée après l'arrêt de la kétamine (1,10 ± 0,17 %; P < 0,05). Les valeurs de MAC avec la kétamine étaient différentes entre les groupes (P < 0,05). Les concentrations plasmatiques de kétamine étaient similaires durant la détermination des MAC.La MAC d'isoflurane durant la CRI de kétamine a donné des résultats différents lorsque les méthodes d'un jour unique (le groupe 1) versus des jours séparés (le groupe 2) étaient utilisées, malgré des concentrations plasmatiques de kétamine similaires avec les deux méthodes. Toutefois, étant donné que l'ampleur de cette différence était de moins de 10 %, chacune des deux méthodes pour déterminer la MAC est considérée comme acceptable à des fins de recherche.(Traduit par Docteur Serge Messier).


Dogs , Isoflurane/pharmacology , Isoflurane/pharmacokinetics , Ketamine/pharmacology , Ketamine/pharmacokinetics , Anesthetics, Dissociative/administration & dosage , Anesthetics, Dissociative/pharmacokinetics , Anesthetics, Dissociative/pharmacology , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/pharmacokinetics , Anesthetics, Inhalation/pharmacology , Animals , Cross-Over Studies , Drug Interactions , Female , Isoflurane/administration & dosage , Ketamine/administration & dosage , Male , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism
20.
Berl Munch Tierarztl Wochenschr ; 127(1-2): 3-11, 2014.
Article De | MEDLINE | ID: mdl-24490337

was observed. Perioperatively oxygen saturation was persistently high and mean arterial pressure was steady, too. An additional Ketamine administration caused a short tachycardia during operation. After restoration of total mobility, respiratory and heart rate stayed within the reference ranges again. All EMG values in between those caused by pain stimuli were significantly below the borderline of a muscle activity in conformity with a clinically visible complete muscle relaxation. Cortisol increased simultaneously with Ketamine and Azaperone before operation, but it remained at this level until the end of the determinations, parallel to the course of Norketamine, close to the maximum before anesthesia. The complex intensive-medical monitoring confirms that under real surgical conditions the counter-regulatory effects of both drugs equalize the respective cardiovascular and respiratory side effects. It is concluded also that the increase of cortisol is likely to be more a side effect of Ketamine/Norketamine than the expression of distress by surgical interventions or by wake-up reactions, and that an intoxication by additional Ketamine dosage or motoric disorders (i.e., catalepsis) can be excluded as undesired side effects of both drugs.


Anesthesia, General/methods , Anesthetics, Dissociative/adverse effects , Azaperone/adverse effects , Hypnotics and Sedatives/adverse effects , Ketamine/adverse effects , Anesthetics, Dissociative/pharmacokinetics , Anesthetics, Dissociative/pharmacology , Anesthetics, Dissociative/therapeutic use , Animals , Azaperone/pharmacokinetics , Azaperone/pharmacology , Azaperone/therapeutic use , Drug Interactions , Electromyography , Heart Rate/drug effects , Hydrocortisone/blood , Hypnotics and Sedatives/pharmacokinetics , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/therapeutic use , Ketamine/pharmacokinetics , Ketamine/pharmacology , Ketamine/therapeutic use , Monitoring, Intraoperative , Respiratory Rate/drug effects , Swine
...