Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
J Clin Lab Anal ; 34(4): e23115, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31733006

ABSTRACT

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.


Subject(s)
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
2.
Pharmacotherapy ; 39(4): 454-462, 2019 04.
Article in English | MEDLINE | ID: mdl-30825333

ABSTRACT

BACKGROUND: Although few studies have used ketamine for induction and maintenance of pediatric anesthesia, official dosage recommendations are lacking. This study evaluates the outcomes of adult anesthetic doses in a pediatric population through pharmacokinetic modeling and computer simulations in an attempt to recommend an adequate ketamine dosing regimen. METHODS: Ketamine plasma concentration-time data in 19 children (age 8 months to 16 years; weight 5.5 to 67 kg) were analyzed according to a non-compartmental pharmacokinetic approach. The relationship between pharmacokinetic parameters and demographic covariates was mathematically characterized. A one-compartment open model was implemented to simulate the plasma profile following administration of 1-4.5 mg/kg IV bolus dose and 0.1-0.5 mg/kg/min continuous infusion of ketamine and to predict anesthesia onset and offset. KEY RESULTS: Pharmacokinetic parameters determined were clearance 0.025 ± 0.008 L/kg/min; distribution volume 3.3 ± 1.3 L/kg; half-life 2.6 ± 1 h; and mean residence time 2.3 ± 0.64 h. Body weight was the best predictor of clearance and distribution volume according to a 0.75-power model. Using weight to scale doses was associated with limited variability in simulated concentrations. Ketamine administered as 2.25 mg/kg IV bolus dose, followed by 0.1 mg/kg/min continuous IV infusion enables anesthesia initiation within 3 minutes and maintains it for 3 hours. CONCLUSIONS & INFERENCES: Weight-based dosing minimizes age-dependent variation in the plasma concentration of ketamine. Low-to-intermediate adult doses are suitable for induction and maintenance of safe anesthesia in children undergoing short-term surgical operations. However, this finding requires validation in controlled clinical trials before it is adopted into surgical standard practices.


Subject(s)
Anesthetics, Dissociative/administration & dosage , Anesthetics, Dissociative/blood , Ketamine/administration & dosage , Ketamine/blood , Models, Biological , Adolescent , Age Factors , Area Under Curve , Body Weight , Child , Child, Preschool , Dose-Response Relationship, Drug , Humans , Infant , Infusions, Intravenous , Injections, Intravenous
3.
Drug Test Anal ; 11(1): 68-76, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30027605

ABSTRACT

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.


Subject(s)
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
4.
Article in English | MEDLINE | ID: mdl-29782960

ABSTRACT

Ketamine hydrochloride elicits potent psychotomimetic and neurobehavioural effects which make it incompatible with driving; however, the direct effect on driving performance is yet to be assessed. Using an open label, within-subjects protocol, 15 males and 5 females (mean age = 30.8 years) were administered three fixed, stepwise increasing sub-anaesthetic doses of intravenous (IV) ketamine solution [(i) 8 mg/h IV infusion plus 30 mg bolus, (ii) 12 mg/h IV infusion and (iii) 20 mg/h infusion]. Whole blood ketamine and norketamine concentrations were determined at each treatment step and at 2 h post-infusion. Driving performance was assessed at baseline, at each treatment step and at 2 h post-treatment using a validated computerised driving simulator. Standard Deviation of Lateral Position (SDLP) and Steering Variability (SV) were assessed. Linear Fixed Effect Modelling indicated a main effect for time (dose) for SDLP (F[4,72] = 33.22, p < 0.0001) and SV (F[4,72] = 4.65, p < 0.002). Post-hoc analyses revealed significant differences from baseline at each treatment step for SDLP (all p < 0.001), and for 12 mg/h treatment step for SV (p = 0.049). Post-treatment driving performance returned to baseline levels. Weak positive linear associations were observed between SDLP and whole blood ketamine concentrations (R2 = 0.11, ß = 29.96, p = 0.001) and norketamine (R2 = 0.09, ß = 28.87, p = 0.003). These findings suggest that even under highly controlled conditions, ketamine intoxication significantly alters simulated driving performance. At the highest dose, ketamine produced changes to SDLP considered incompatible with safe driving, highlighting how ketamine consumption may translate to an increased risk of road trauma.


Subject(s)
Anesthetics, Dissociative/administration & dosage , Automobile Driving , Ketamine/administration & dosage , Adult , Anesthetics, Dissociative/blood , Computer Simulation , Dose-Response Relationship, Drug , Female , Humans , Ketamine/analogs & derivatives , Ketamine/blood , Male , Psychomotor Performance/drug effects , Young Adult
5.
Basic Clin Pharmacol Toxicol ; 122(5): 481-488, 2018 May.
Article in English | MEDLINE | ID: mdl-29171155

ABSTRACT

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.


Subject(s)
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
6.
Forensic Sci Int ; 267: e1-e5, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27616733

ABSTRACT

Animal cruelty is a known behavior of psychopaths, and although the serial killing of humans is widely acknowledged worldwide, this type of crime against animals is seldom discussed. This report describes the necropsy and toxicological findings of 37 dogs and cats, which were found dead in plastic bags in Sao Paulo, Brazil. The animals had all been in the care of an alleged animal rescuer and were to be referred for adoption before being found dead. In the necropsy, the animals showed varying degrees of putrefaction, indicating different periods of death, as well as single or multiple perforations on the thorax. The perforations reached the heart, lungs or large thoracic vessels, culminating in hemopericardium and hemothorax that led to death by circulatory failure and cardiac tamponade. Blood from the heart and thoracic cavity was analyzed by gas chromatography coupled with mass spectrometry (GC-MS) and tested positive for ketamine, a dissociative anesthetic. The suspect declared that she had killed only five of the animals and that they had all been fatally sick. The necropsy proved that all 37 animals were killed in the same way, that none of the animals had any terminal diseases and that a restricted drug was used. The suspect was sentenced to 12 years, 6 months and 14days of prison for the killing of the 37 animals. This was the first conviction for the crime of animal cruelty in Brazil. The combined role of police, forensic veterinary pathologists and prosecutors were essential to the conviction, which was a great historical occasion in the fight against animal cruelty.


Subject(s)
Anesthetics, Dissociative/blood , Animal Welfare/legislation & jurisprudence , Crime , Ketamine/blood , Animals , Brazil , Cats , Dogs , Female , Gas Chromatography-Mass Spectrometry , Humans
7.
Forensic Sci Int ; 265: 193-9, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27017567

ABSTRACT

Ketamine (KT) is used to induce and maintain general anaesthesia in combination with sedative drugs in human and animals. Because of its dissociative and hallucinogenic effects, KT has become a recreational drug in a variety of social settings and may be included in the panel of drugs of abuse that are controlled in driving under the influence (DUI) ascertainment. In a local driving license re-granting protocol, a case where a veterinary physician was found positive to KT and nor-ketamine (NK) in hair suggested the possibility of a professional exposure in a veterinary setting and prompted an experimental study. Male (7) and female (4) veterinary physicians were recruited on a voluntary base. Detailed information was collected on their habits, use of drugs, professional practice, frequency and mode of using KT injections. Hands and skin were examined. Head hair and pubic hair were collected. Two naïve subjects, starting their professional practice at a local veterinary clinic, were recruited and their hair (head, pubic, axillary, thoracic hair, and beard) and urine were collected before and after usual clinic activity. Hair were cut according to their length, washed, pulverized and 25mg were extracted and analyzed by liquid chromatography coupled to high accuracy, high resolution mass spectrometry. All the hair samples from the veterinary physicians turned to be positive for KT, at a concentration varying from 0.010 to 0.840ng/mg in head hair and from 0.040 to 2.04ng/mg in pubic hair; NK ranged from not detected to 0.080ng/mg in head hair, from not detected to 0.100 in pubic hair; when KT was ≥0.100, NK was always detected. For the two naïve subjects, hair from different body sites were negative before they started their activity, and positive one month later; some urine samples resulted positive and confirmed systemic exposure to KT. The possibility of unaware exposure to KT was demonstrated. The site of absorption is skin, independently from the presence of skin injuries due to hand contact with KT injection solutions and/or animal body fluids by veterinary physicians during clinical activities and animal handling. Possible adverse systemic effects by unaware KT exposure need to be studied and concerns have arisen as to healthy and safe workplace policies. Comparison with a population of subjects undergoing driving license re-granting evidenced a larger range of concentration in this latter setting (0.050-10.0ng/mg for KT, not detected to 0.100ng/mg for NK).


Subject(s)
Anesthetics, Dissociative/blood , Hair/chemistry , Ketamine/blood , Occupational Exposure , Adult , Female , Humans , Male , Middle Aged , Prospective Studies , Retrospective Studies , Veterinary Medicine , Young Adult
8.
Int J Legal Med ; 130(2): 405-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26482953

ABSTRACT

Methoxydiphenidine (MXP) was first reported in 1989 as a dissociative anesthetic but did not enter the market for pharmaceuticals. The substance re-appeared in 2013 as a new psychoactive substance. A case of driving under the influence of MXP is reported. The concentration of MXP has been determined from a serum sample (57 ng/mL) by liquid chromatography tandem mass spectrometry following liquid-liquid extraction. In addition, amphetamine, methylenedioxymethamphetamine, and its major metabolite were present in concentrations of 111, 28, and 3 ng/mL, respectively. The subject presented with amnesia, out-of-body experiences, bizarre behavior, and decreased motor abilities. At present, information on human toxicity of MXP is not available. MXP is comparable in structure as well as in action at the N-methyl-D-aspartate (NMDA) receptor to phencyclidine or ketamine. Therefore, it is likely that MXP exerts similar severe psychotropic action in man. However, there is no information on the duration and intensity of MXP's impairing effects, the interpretation of a particular concentration in the blood or serum, and its detectability in routine drug screenings. Confirmation analysis may be confined to cases where the police has specific intelligence that points to MXP use.


Subject(s)
Anesthetics, Dissociative/adverse effects , Driving Under the Influence , Piperidines/adverse effects , Adult , Anesthetics, Dissociative/blood , Chromatography, Liquid , Humans , Male , Mass Spectrometry , Piperidines/blood , Substance-Related Disorders/blood , Substance-Related Disorders/complications
9.
J Anal Toxicol ; 39(4): 287-93, 2015 May.
Article in English | MEDLINE | ID: mdl-25698777

ABSTRACT

2-Methoxydiphenidine, i.e. 1-[1-(2-methoxyphenyl)-2-phenylethyl]piperidine, also known as 'MXP' or '2-MeO-diphenidine' (or 2-MXP), has been available as a 'research chemical' since 2013 as a purported alternative to the 'dissociative anesthetics' methoxetamine and ketamine. Three deaths which involved the detection of 2-MXP in post-mortem blood and urine were encountered in forensic casework. The 2-, 3- and 4-methoxyphenyl positional isomers were synthesized to confirm the identity and concentration of 2-MXP. The 2-MXP femoral blood concentrations in the cases were found to be 24.0, 2.0 and 1.36 mg/L (the latter with an alternative cause of death). Some additional prescription drugs were encountered at therapeutic concentrations in all three cases. Analysis of the biofluids allowed the detection and characterization of various metabolites, including the suggested presence of hydroxy-2-MXP as the main metabolite with the hydroxyl group located on the piperidine rather than the phenyl or benzyl moiety. Additional metabolites included O-desmethyl-2-MXP and hydroxylated O-desmethyl-2-MXP. Diphenidine and hydroxy-diphenidine, also showing the presence of the hydroxyl group on the piperidine ring, were also detected. It was not possible to identify whether these arose from 2-MXP biotransformation or whether they represented the presence of diphenidine as a separate substance. These are the first published fatalities involving 2-MXP and presents analytical data to assist analytical toxicologists with future casework.


Subject(s)
Anesthetics, Dissociative/blood , Anesthetics, Dissociative/poisoning , Piperidines/blood , Piperidines/poisoning , Adult , Fatal Outcome , Humans , Male , Poisoning/blood
10.
Paediatr Anaesth ; 25(2): 211-6, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25212712

ABSTRACT

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.


Subject(s)
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
11.
Int J Pharm ; 478(1): 223-231, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25448584

ABSTRACT

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).


Subject(s)
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
13.
J Am Assoc Lab Anim Sci ; 52(5): 567-70, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24041212

ABSTRACT

To compare the pharmacokinetics of coadministered intraperitoneal ketamine and xylazine in young (8 to 10 wk; n = 6) and old rats (2 to 2.4 y; n = 6), blood samples obtained at 15 and 30 min and 1, 2, and 4 h after drug administration were analyzed by HPLC-tandem mass spectrometry. In both groups, the withdrawal reflex was absent during anesthesia and was present at 1.1 (± 0.2) and 2.6 (± 0.7) h after drug administration in young and old rats, respectively, with the first voluntary movement at 1.5 ± 0.2 and 4.9 ± 1.0 h. Drug availability of ketamine and xylazine was 6.0 and 6.7 times greater, respectively, in old than young rats. The rate constant of elimination of both drugs was greatly decreased and the elimination half-life was significantly greater in old compared with young rats. In conclusion, age and associated factors affect the availability of ketamine and xylazine when coadministered to attain clinical anesthesia, changing the pharmacokinetics of these drugs and prolonging anesthesia duration and recovery times with aging. Compared with their young counterparts, aged rats required much higher doses to attain a similar level of anesthesia. Finally, the long half-life of both ketamine and xylazine, when coadministered to old rats, may be a factor in research protocols because residual plasma concentrations could still be present for as long as 3 and 5 d, respectively, after administration.


Subject(s)
Anesthesia/veterinary , Anesthetics, Dissociative/pharmacokinetics , Hypnotics and Sedatives/pharmacokinetics , Ketamine/pharmacokinetics , Xylazine/pharmacokinetics , Age Factors , Aging , Anesthesia/methods , Anesthetics, Dissociative/blood , Animals , Drug Therapy, Combination , Humans , Hypnotics and Sedatives/blood , Ketamine/blood , Rats , Rats, Sprague-Dawley , Xylazine/blood
14.
J Chromatogr A ; 1264: 104-9, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23058940

ABSTRACT

A new method was developed for determination of ketamine (KT) in urine and plasma samples by derivatization and gas chromatography-mass spectrometry. In this article, KT was first derivatized with sodium nitrite to volatile N-nitrosamines under acidic condition. Then the derivative had been identified by the mass spectra. The derivatization conditions including the amount of hydrochloric acid, the amount of sodium nitrite, reaction temperature, reaction time and the extraction reagents were optimized. Calibration curves were linear in the range of 0.04-20 µg mL(-1) for KT, and the limit of detection (LOD) and limit of quantitation (LOQ) were 0.01 µg mL(-1) and 0.04 µg mL(-1), respectively. The results of recovery indicated that the method had good precision and reproducibility. Compared with existing derivatization methods, this method provided a rapid, convenient, effective and low-cost way for gas chromatography method of KT quantification.


Subject(s)
Anesthetics, Dissociative/blood , Anesthetics, Dissociative/urine , Gas Chromatography-Mass Spectrometry/methods , Ketamine/blood , Ketamine/urine , Humans , Limit of Detection
15.
Pediatr Emerg Care ; 28(8): 767-70, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22858745

ABSTRACT

OBJECTIVE: The objective of this study was to compare the efficacy of 3 doses of intranasal ketamine (INK) for sedation of children from 1 to 7 years old requiring laceration repair. METHODS: This was a randomized, prospective, double-blind trial of children requiring sedation for laceration repair. Patients with simple lacerations were randomized by age to receive 3, 6, or 9 mg/kg INK. Adequacy and efficacy of sedation were measured with the Ramsay sedation score and the Observational Scale of Behavioral Distress-Revised. Serum ketamine and norketamine levels were drawn during the procedure. Sedation duration and adverse events were recorded. RESULTS: Of the 12 patients enrolled, 3 patients achieved adequate sedation, all at the 9-mg/kg dose. The study was suspended at that time as per predetermined criteria. CONCLUSIONS: Nine milligrams of INK per kilogram produced a significantly higher proportion of successful sedations than the 3- and 6-mg/kg doses.


Subject(s)
Anesthetics, Dissociative/administration & dosage , Ketamine/administration & dosage , Lacerations/surgery , Administration, Intranasal , Anesthetics, Dissociative/blood , Child , Child, Preschool , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Infant , Ketamine/analogs & derivatives , Ketamine/blood , Prospective Studies
16.
Anesthesiology ; 117(4): 868-77, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22890117

ABSTRACT

BACKGROUND: The influence of psychoactive drugs on the central nervous system has been investigated with positron emission tomography and task-related functional magnetic resonance imaging. However, it is not known how these drugs affect the intrinsic large-scale interactions of the brain (resting-state functional magnetic resonance imaging connectivity). In this study, the effect of low-dose S(+)-ketamine on intrinsic brain connectivity was investigated. METHODS: Twelve healthy, male volunteers received a 2-h intravenous S(+)-ketamine infusion (first hour 20 mg/70 kg, second hour 40 mg/70 kg). Before, during, and after S(+)-ketamine administration, resting-state brain connectivity was measured. In addition, heat pain tests were performed between imaging sessions to determine ketamine-induced analgesia. A mixed-effects general linear model was used to determine drug and pain effects on resting-state brain connectivity. RESULTS: Ketamine increased the connectivity most importantly in the cerebellum and visual cortex in relation to the medial visual network. A decrease in connectivity was observed in the auditory and somatosensory network in relation to regions responsible for pain sensing and the affective processing of pain, which included the amygdala, insula, and anterior cingulate cortex. Connectivity variations related to fluctuations in pain scores were observed in the anterior cingulate cortex, insula, orbitofrontal cortex, and the brainstem, regions involved in descending inhibition of pain. CONCLUSIONS: Changes in connectivity were observed in the areas that explain ketamine's pharmacodynamic profile with respect to analgesia and psychedelic and other side effects. In addition, pain and ketamine changed brain connectivity in areas involved in endogenous pain modulation.


Subject(s)
Anesthetics, Dissociative/pharmacology , Brain/drug effects , Ketamine/pharmacology , Magnetic Resonance Imaging , Nerve Net/anatomy & histology , Nerve Net/drug effects , Adult , Anesthetics, Dissociative/adverse effects , Anesthetics, Dissociative/blood , Data Interpretation, Statistical , Hallucinogens/pharmacology , Hot Temperature , Humans , Image Processing, Computer-Assisted , Injections, Intravenous , Ketamine/adverse effects , Ketamine/analogs & derivatives , Ketamine/blood , Linear Models , Male , Pain/psychology , Pain Measurement/drug effects , Single-Blind Method , Young Adult
17.
J Psychopharmacol ; 26(9): 1211-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22389244

ABSTRACT

Evidence suggests that some aspects of schizophrenia can be induced in healthy volunteers through acute administration of the non-competitive NMDA-receptor antagonist, ketamine. In probabilistic inference tasks, patients with schizophrenia have been shown to 'jump to conclusions' (JTC) when asked to make a decision. We aimed to test whether healthy participants receiving ketamine would adopt a JTC response pattern resembling that of patients. The paradigmatic task used to investigate JTC has been the 'urn' task, where participants are shown a sequence of beads drawn from one of two 'urns', each containing coloured beads in different proportions. Participants make a decision when they think they know the urn from which beads are being drawn. We compared performance on the urn task between controls receiving acute ketamine or placebo with that of patients with schizophrenia and another group of controls matched to the patient group. Patients were shown to exhibit a JTC response pattern relative to their matched controls, whereas JTC was not evident in controls receiving ketamine relative to placebo. Ketamine does not appear to promote JTC in healthy controls, suggesting that ketamine does not affect probabilistic inferences.


Subject(s)
Anesthetics, Dissociative/adverse effects , Decision Making/drug effects , Excitatory Amino Acid Antagonists/adverse effects , Ketamine/adverse effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Schizophrenia/physiopathology , Schizophrenic Psychology , Adult , Anesthetics, Dissociative/administration & dosage , Anesthetics, Dissociative/blood , Antipsychotic Agents/therapeutic use , Diagnostic and Statistical Manual of Mental Disorders , Dose-Response Relationship, Drug , Double-Blind Method , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/blood , Female , Humans , Ketamine/administration & dosage , Ketamine/blood , Male , Middle Aged , Neurotoxicity Syndromes/diagnosis , Patient Dropouts , Schizophrenia/blood , Schizophrenia/chemically induced , Schizophrenia/drug therapy , Severity of Illness Index , Task Performance and Analysis , Young Adult
18.
Vet Anaesth Analg ; 38(4): 292-300, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21627754

ABSTRACT

OBJECTIVE: To determine the effect of intravenous ketamine on the minimum alveolar concentration of sevoflurane needed to block autonomic response (MAC(BAR)) to a noxious stimulus in dogs. STUDY DESIGN: Randomized, crossover, prospective design. ANIMALS: Eight, healthy, adult male, mixed-breed dogs, weighing 11.2-16.1 kg. METHODS: Dogs were anesthetized with sevoflurane on two occasions, 1 week apart, and baseline MAC(BAR) (B-MAC(BAR)) was determined on each occasion. MAC(BAR) was defined as the mean of the end-tidal sevoflurane concentrations that prevented and allowed an increase (≥15%) in heart rate or invasive mean arterial pressure in response to a noxious electrical stimulus (50 V, 50 Hz, 10 ms). Dogs then randomly received either a low-dose (LDS) or high-dose series (HDS) of ketamine, and treatment MAC(BAR) (T-MAC(BAR)) was determined. The LDS had an initial loading dose (LD) of 0.5 mg kg(-1) and constant rate infusion (CRI) at 6.25 µg kg(-1) minute(-1), followed, after T-MAC(BAR) determination, by a second LD (1 mg kg(-1)) and CRI (12.5 µg kg(-1) minute(-1)). The HDS had an initial LD (2 mg kg(-1)) and CRI (25 µg kg(-1) minute(-1)) followed by a second LD (3 mg kg(-1)) and CRI (50 µg kg(-1) minute(-1)). Data were analyzed with a mixed-model anova and are presented as LSM ± SEM. RESULTS: The B-MAC(BAR) was not significantly different between treatments. Ketamine at 12.5, 25, and 50 µg kg(-1) minute(-1) decreased sevoflurane MAC(BAR), and the maximal decrease (22%) occurred at 12.5 µg kg(-1) minute(-1). The percentage change in MAC(BAR) was not correlated with either the log plasma ketamine or norketamine concentration. CONCLUSIONS AND CLINICAL RELEVANCE: Ketamine at clinically relevant doses of 12.5, 25, and 50 µg kg(-1) minute(-1) decreased sevoflurane MAC(BAR), although the reduction was neither dose-dependent nor linear.


Subject(s)
Anesthesia, Inhalation/veterinary , Anesthesia, Intravenous/veterinary , Anesthetics, Dissociative/pharmacology , Anesthetics, Inhalation/pharmacology , Autonomic Nervous System/drug effects , Dogs , Ketamine/pharmacology , Methyl Ethers/pharmacology , Anesthesia Recovery Period , Anesthetics, Dissociative/blood , Animals , Blood Pressure/drug effects , Cross-Over Studies , Dose-Response Relationship, Drug , Heart Rate/drug effects , Ketamine/blood , Male , Prospective Studies , Pulmonary Alveoli , Sevoflurane
19.
Anesthesiology ; 114(6): 1435-45, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21508826

ABSTRACT

BACKGROUND: Low-dose ketamine is used as analgesic for acute and chronic pain. It is metabolized in the liver to norketamine via cytochrome P450 (CYP) enzymes. There are few human data on the involvement of CYP enzymes on the elimination of norketamine and its possible contribution to analgesic effect. The aim of this study was to investigate the effect of CYP enzyme induction by rifampicin on the pharmacokinetics of S-ketamine and its major metabolite, S-norketamine, in healthy volunteers. METHODS: Twenty healthy male subjects received 20 mg/70 kg/h (n = 10) or 40 mg/70 kg/h (n = 10) intravenous S-ketamine for 2 h after either 5 days oral rifampicin (once daily 600 mg) or placebo treatment. During and 3 h after drug infusion, arterial plasma concentrations of S-ketamine and S-norketamine were obtained at regular intervals. The data were analyzed with a compartmental pharmacokinetic model consisting of three compartments for S-ketamine, three sequential metabolism compartments, and two S-norketamine compartments using the statistical package NONMEM® 7 (ICON Development Solutions, Ellicott City, MD). RESULTS: Rifampicin caused a 10% and 50% reduction in the area-under-the-curve of the plasma concentrations of S-ketamine and S-norketamine, respectively. The compartmental analysis indicated a 13% and 200% increase in S-ketamine and S-norketamine elimination from their respective central compartments by rifampicin. CONCLUSIONS: : A novel observation is the large effect of rifampicin on S-norketamine concentrations and indicates that rifampicin induces the elimination of S-ketamine's metabolite, S-norketamine, probably via induction of the CYP3A4 and/or CYP2B6 enzymes.


Subject(s)
Ketamine/analogs & derivatives , Ketamine/blood , Rifampin/pharmacology , Adult , Anesthetics, Dissociative/administration & dosage , Anesthetics, Dissociative/blood , Cross-Over Studies , Cytochrome P-450 Enzyme System/biosynthesis , Drug Interactions/physiology , Enzyme Induction/drug effects , Enzyme Induction/physiology , Humans , Infusions, Intravenous , Ketamine/administration & dosage , Male , Single-Blind Method
20.
Biomed Chromatogr ; 25(12): 1374-80, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21381066

ABSTRACT

Ketamine and medetomidine are commonly combined to sedate or anaesthetize a wide range of animal species. Despite this, there are few methods for the simultaneous quantitative analysis of the two drugs. This study describes the use of solid-phase extraction sample preparation followed by liquid chromatography-tandem mass spectrometry for the quantitative analysis of both drugs in ovine plasma. Extraction recovery was 93% for ketamine and 95% for medetomidine. The lowest limit of detection for ketamine was 1 ng/mL and for medetomidine 2 ng/mL, with linearity greater than 0.99 for both. Intra-day and inter-day precisions for both drugs were less than 10 and 7%, respectively. Application of the method to samples obtained from pregnant ewes and their fetuses showed placental transfer of the drugs over time such that there was no significant difference in plasma concentration at delivery. In summary, a validated method has been developed for the simultaneous quantification of ketamine and medetomidine in ovine plasma samples which can be used to study the pharmacokinetics of these drugs.


Subject(s)
Chromatography, Liquid/methods , Ketamine/blood , Medetomidine/blood , Tandem Mass Spectrometry/methods , Anesthetics, Dissociative/blood , Anesthetics, Dissociative/pharmacokinetics , Animals , Animals, Newborn , Female , Ketamine/pharmacokinetics , Maternal-Fetal Exchange , Medetomidine/pharmacokinetics , Pregnancy , Reproducibility of Results , Sensitivity and Specificity , Sheep , Solid Phase Extraction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...