Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 315
Filtrar
1.
Toxicology ; 500: 153682, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38006927

RESUMEN

Ketamine is a psychotropic drug that can cause significant neurological symptoms and is closely linked to the activity of the CYP3A4 enzyme. This study aimed to examine the diversity of CYP3A4 activity affects the metabolism of ketamine, focusing on genetic variation and drug-induced inhibition. We used a baculovirus-insect cell expression system to prepare recombinant human CYP3A4 microsomes. Then, in vitro enzyme incubation systems were established and used UPLC-MS/MS to detect ketamine metabolite. In rats, we investigated the metabolism of ketamine and its metabolite in the presence of the CYP3A4 inhibitor voriconazole. Molecular docking was used to explore the molecular mechanism of inhibition. The results showed that the catalytic activity of CYP3A4.5, .17, .23, .28, and .29 significantly decreased compared to CYP3A4.1, with a minimum decrease of 3.13%. Meanwhile, the clearance rate of CYP3A4.2, .32, and .34 enhanced remarkably, ranging from 40.63% to 87.50%. Additionally, hepatic microsome incubation experiments revealed that the half-maximal inhibitory concentration (IC50) of voriconazole for ketamine in rat and human liver microsomes were 18.01 ± 1.20 µM and 14.34 ± 1.70 µM, respectively. When voriconazole and ketamine were co-administered, the blood exposure of ketamine and norketamine significantly increased in rats, as indicated by the area under the concentration-time curve (AUC) and maximum concentration (Cmax). The elimination half-life (t1/2Z) of these substances was also prolonged. Moreover, the clearance (CLz/F) of ketamine decreased, while the apparent volume of distribution (Vz/F) increased significantly. This might be attributed to the competition between voriconazole and ketamine for binding sites on the CYP3A4 enzyme. In conclusion, variations in CYP3A4 activity would result in the stratification of ketamine blood exposure.


Asunto(s)
Citocromo P-450 CYP3A , Ketamina , Animales , Humanos , Ratas , Cromatografía Liquida , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Ketamina/metabolismo , Ketamina/farmacocinética , Microsomas Hepáticos/metabolismo , Simulación del Acoplamiento Molecular , Espectrometría de Masas en Tándem , Voriconazol/metabolismo , Voriconazol/farmacología
2.
Nature ; 622(7984): 802-809, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37853123

RESUMEN

Ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist1, has revolutionized the treatment of depression because of its potent, rapid and sustained antidepressant effects2-4. Although the elimination half-life of ketamine is only 13 min in mice5, its antidepressant activities can last for at least 24 h6-9. This large discrepancy poses an interesting basic biological question and has strong clinical implications. Here we demonstrate that after a single systemic injection, ketamine continues to suppress burst firing and block NMDARs in the lateral habenula (LHb) for up to 24 h. This long inhibition of NMDARs is not due to endocytosis but depends on the use-dependent trapping of ketamine in NMDARs. The rate of untrapping is regulated by neural activity. Harnessing the dynamic equilibrium of ketamine-NMDAR interactions by activating the LHb and opening local NMDARs at different plasma ketamine concentrations, we were able to either shorten or prolong the antidepressant effects of ketamine in vivo. These results provide new insights into the causal mechanisms of the sustained antidepressant effects of ketamine. The ability to modulate the duration of ketamine action based on the biophysical properties of ketamine-NMDAR interactions opens up new opportunities for the therapeutic use of ketamine.


Asunto(s)
Antidepresivos , Depresión , Habénula , Ketamina , Receptores de N-Metil-D-Aspartato , Animales , Ratones , Antidepresivos/administración & dosificación , Antidepresivos/metabolismo , Antidepresivos/farmacocinética , Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Depresión/metabolismo , Habénula/efectos de los fármacos , Habénula/metabolismo , Semivida , Ketamina/administración & dosificación , Ketamina/metabolismo , Ketamina/farmacocinética , Ketamina/farmacología , Neuronas/fisiología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Factores de Tiempo , Unión Proteica
3.
Br J Pharmacol ; 179(1): 65-83, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34519023

RESUMEN

BACKGROUND AND PURPOSE: Deschloroketamine (DCK), a structural analogue of ketamine, has recently emerged on the illicit drug market as a recreational drug with a modestly long duration of action. Despite it being widely used by recreational users, no systematic research on its effects has been performed to date. EXPERIMENTAL APPROACH: Pharmacokinetics, acute effects, and addictive potential in a series of behavioural tests in Wistar rats were performed following subcutaneous (s.c.) administration of DCK (5, 10, and 30 mg·kg-1 ) and its enantiomers S-DCK (10 mg·kg-1 ) and R-DCK (10 mg·kg-1 ). Additionally, activity at human N-methyl-d-aspartate (NMDA) receptors was also evaluated. KEY RESULTS: DCK rapidly crossed the blood brain barrier, with maximum brain levels achieved at 30 min and remaining high at 2 h after administration. Its antagonist activity at NMDA receptors is comparable to that of ketamine with S-DCK being more potent. DCK had stimulatory effects on locomotion, induced place preference, and robustly disrupted PPI. Locomotor stimulant effects tended to disappear more quickly than disruptive effects on PPI. S-DCK had more pronounced stimulatory properties than its R-enantiomer. However, the potency in disrupting PPI was comparable in both enantiomers. CONCLUSION AND IMPLICATIONS: DCK showed similar behavioural and addictive profiles and pharmacodynamics to ketamine, with S-DCK being in general more active. It has a slightly slower pharmacokinetic profile than ketamine, which is consistent with its reported longer duration of action. These findings have implications and significance for understanding the risks associated with illicit use of DCK.


Asunto(s)
Conducta Animal , Drogas Ilícitas , Ketamina , Locomoción , Animales , Conducta Animal/efectos de los fármacos , Drogas Ilícitas/efectos adversos , Drogas Ilícitas/farmacocinética , Drogas Ilícitas/farmacología , Ketamina/administración & dosificación , Ketamina/efectos adversos , Ketamina/análogos & derivados , Ketamina/farmacocinética , Ketamina/farmacología , Locomoción/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/metabolismo
4.
J Psychopharmacol ; 36(2): 170-182, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34971525

RESUMEN

BACKGROUND: Ketamine is rapidly metabolized to norketamine and hydroxynorketamine (HNK) metabolites. In female mice, when compared to males, higher levels of (2R,6R;2S,6S)-HNK have been observed following ketamine treatment, and higher levels of (2R,6R)-HNK following the direct administration of (2R,6R)-HNK. AIM: The objective of this study was to evaluate the impact of sex in humans and mice, and gonadal hormones in mice on the metabolism of ketamine to form norketamine and HNKs and in the metabolism/elimination of (2R,6R)-HNK. METHODS: In CD-1 mice, we utilized gonadectomy to evaluate the role of circulating gonadal hormones in mediating sex-dependent differences in ketamine and (2R,6R)-HNK metabolism. In humans (34 with treatment-resistant depression and 23 healthy controls) receiving an antidepressant dose of ketamine (0.5 mg/kg i.v. infusion over 40 min), we evaluated plasma levels of ketamine, norketamine, and HNKs. RESULTS: In humans, plasma levels of ketamine and norketamine were higher in males than females, while (2R,6R;2S,6S)-HNK levels were not different. Following ketamine administration to mice (10 mg/kg i.p.), Cmax and total plasma concentrations of ketamine and norketamine were higher, and those of (2R,6R;2S,6S)-HNK were lower, in intact males compared to females. Direct (2R,6R)-HNK administration (10 mg/kg i.p.) resulted in higher levels of (2R,6R)-HNK in female mice. Ovariectomy did not alter ketamine metabolism in female mice, whereas orchidectomy recapitulated female pharmacokinetic differences in male mice, which was reversed with testosterone replacement. CONCLUSION: Sex is an important biological variable that influences the metabolism of ketamine and the HNKs, which may contribute to sex differences in therapeutic antidepressant efficacy or side effects.


Asunto(s)
Antidepresivos/farmacocinética , Trastorno Depresivo Resistente al Tratamiento/tratamiento farmacológico , Ketamina/farmacocinética , Adulto , Animales , Antidepresivos/administración & dosificación , Estudios de Casos y Controles , Estudios Cruzados , Femenino , Humanos , Ketamina/administración & dosificación , Ketamina/análogos & derivados , Masculino , Ratones , Persona de Mediana Edad , Orquiectomía , Ovariectomía , Factores Sexuales , Especificidad de la Especie , Adulto Joven
5.
Neuropharmacology ; 206: 108936, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-34965407

RESUMEN

Esketamine, the S-stereoisomer of (R,S)-ketamine was recently approved by drug agencies (FDA, EMA), as an antidepressant drug with a new mechanism of action. (R,S)-ketamine is a N-methyl-d-aspartate receptor (NMDA-R) antagonist putatively acting on GABAergic inhibitory synapses to increase excitatory synaptic glutamatergic neurotransmission. Unlike monoamine-based antidepressants, (R,S)-ketamine exhibits rapid and persistent antidepressant activity at subanesthetic doses in preclinical rodent models and in treatment-resistant depressed patients. Its major brain metabolite, (2R,6R)-hydroxynorketamine (HNK) is formed following (R,S)-ketamine metabolism by various cytochrome P450 enzymes (CYP) mainly activated in the liver depending on routes of administration [e.g., intravenous (largely used for a better bioavailability), intranasal spray, intracerebral, subcutaneous, intramuscular or oral]. Experimental or clinical studies suggest that (2R,6R)-HNK could be an antidepressant drug candidate. However, questions still remain regarding its molecular and cellular targets in the brain and its role in (R,S)-ketamine's fast-acting antidepressant effects. The purpose of the present review is: 1) to review (R,S)-ketamine pharmacokinetic properties in humans and rodents and its metabolism by CYP enzymes to form norketamine and HNK metabolites; 2) to provide a summary of preclinical strategies challenging the role of these metabolites by modifying (R,S)-ketamine metabolism, e.g., by administering a pre-treatment CYP inducers or inhibitors; 3) to analyze the influence of sex and age on CYP expression and (R,S)-ketamine metabolism. Importantly, this review describes (R,S)-ketamine pharmacodynamics and pharmacokinetics to alert clinicians about possible drug-drug interactions during a concomitant administration of (R,S)-ketamine and CYP inducers/inhibitors that could enhance or blunt, respectively, (R,S)-ketamine's therapeutic antidepressant efficacy in patients.


Asunto(s)
Antidepresivos/farmacocinética , Sistema Enzimático del Citocromo P-450/metabolismo , Ketamina/análogos & derivados , Animales , Humanos , Ketamina/farmacocinética
6.
Drug Test Anal ; 13(9): 1689-1692, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34121336

RESUMEN

Ketamine is a widely used dissociative drug, whose quantification in plasma and urine can be of pharmacological, toxicological, and clinical interest. Although tandem mass spectrometry allows the reliable determination of ketamine and its metabolites in biological matrices, the structural similarity between norketamine (main active metabolite) and dehydronorketamine (a less relevant metabolite) can represent a critical aspect. These compounds differ exclusively in two hydrogen atoms, but the consequent two-unit difference in their mass/charge ratio is partially nullified by the isotopic abundance of the chlorine atom present in their structure. This, along with their similar fragmentation pattern, can result in the incorrect identification of the enantiomers of these ketamine metabolites even with triple quadrupole instruments, if shared transitions are monitored after chiral chromatography. The key to prevent norketamine overestimation is therefore observing analyte-specific MS/MS transitions. Here, we describe in detail how we investigated this issue, during the development of an analytical method for ketamine and norketamine enantiomer determination in plasma.


Asunto(s)
Cromatografía Liquida/métodos , Ketamina/análogos & derivados , Ketamina/análisis , Espectrometría de Masas en Tándem/métodos , Animales , Perros , Ketamina/sangre , Ketamina/farmacocinética , Estereoisomerismo
7.
J Clin Pharmacol ; 61(11): 1421-1429, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34125442

RESUMEN

Patients with refractory chronic migraine have substantial disability and have failed many acute and preventive medications. When aggressive intravenous therapy is indicated, both lidocaine and (R,S)-ketamine infusions have been used successfully to provide relief. Retrospective studies have shown that both agents may be associated with short-term analgesia. In this prospective, observational pilot study of 6 patients, we compared the effects of lidocaine and (R,S)-ketamine infusions and performed metabolite analyses of (R,S)-ketamine to determine its metabolic profile in this population. One of (R,S)-ketamine's metabolites, (2R,6R)-hydroxynorketamine, has been shown in animal studies to reduce pain, but human studies in patients undergoing continuous (R,S)-ketamine infusions for migraine are lacking. All 6 patients tolerated both infusions well with mild adverse effects. The baseline mean pain rating (0-10 numeric rating scale) decreased from 7.5 ± 2.2 to 4.7 ± 2.8 by end of lidocaine treatment ( P≤.05 ) but increased to 7.0 ± 1.4 by the postdischarge visit at 4 weeks (P > .05 vs baseline). The baseline mean pain rating prior to ketamine treatment was 7.4 ± 1.4, which decreased to 3.7 ± 2.3 by the end of the hospitalization ( P≤.05 ) but increased to 7.2 ± 1.7 by the postdischarge visit at 6 weeks (P > .05 vs baseline). For the primary outcome the change in pain from baseline to end of treatment was greater for ketamine than lidocaine (-3.7 vs -2.8; P≤.05 ), but this has minimal clinical significance. Ketamine metabolite analysis revealed that (2R,6R)-hydroxynorketamine was the predominant metabolite during most of the infusion, consistent with previous studies.


Asunto(s)
Analgésicos/uso terapéutico , Ketamina/uso terapéutico , Lidocaína/uso terapéutico , Trastornos Migrañosos/tratamiento farmacológico , Adulto , Analgésicos/administración & dosificación , Analgésicos/efectos adversos , Enfermedad Crónica , Femenino , Humanos , Ketamina/administración & dosificación , Ketamina/efectos adversos , Ketamina/análogos & derivados , Ketamina/sangre , Ketamina/farmacocinética , Lidocaína/administración & dosificación , Lidocaína/efectos adversos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Estudios Prospectivos , Adulto Joven
8.
Orthop Nurs ; 40(3): 189-193, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34004619

RESUMEN

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.


Asunto(s)
Analgesia , Anestésicos Disociativos , Ketamina , Manejo del Dolor , Dolor Postoperatorio/tratamiento farmacológico , Analgésicos/administración & dosificación , Analgésicos Opioides/uso terapéutico , Anestésicos Disociativos/administración & dosificación , Anestésicos Disociativos/farmacocinética , Dolor en Cáncer/tratamiento farmacológico , Humanos , Ketamina/administración & dosificación , Ketamina/farmacocinética , Dimensión del Dolor
9.
Anesthesiology ; 135(2): 326-339, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34019627

RESUMEN

BACKGROUND: The anesthetic ketamine after intravenous dosing is nearly completely metabolized to R- and S-stereoisomers of the active norketamine (analgesic, psychoactive) and 2,6-hydroxynorketamine (potential analgesic, antidepressant) as well as the inactive dehydronorketamine. Oral administration favors the formation of 2,6-hydroxynorketamines via extensive presystemic metabolism. The authors hypothesized that plasma exposure to 2,6-hydroxynorketamines relative to the psychoactive ketamine is greater after prolonged-release ketamine tablets than it is after intravenous ketamine. METHODS: Pharmacokinetics of ketamine after intravenous infusion (5.0 mg) and single-dose administrations of 10, 20, 40, and 80 mg prolonged-released tablets were evaluated in 15 healthy white human subjects by means of a controlled, ascending-dose study. The stereoisomers of ketamine and metabolites were quantified in serum and urine by validated tandem mass-spectrometric assays and evaluated by noncompartmental pharmacokinetic analysis. RESULTS: After 40 mg prolonged-release tablets, the mean ± SD area under the concentrations-time curve ratios for 2,6-hydroxynorketamine/ketamine were 18 ± 11 (S-stereoisomers) and 30 ± 16 (R-stereoisomers) compared to 1.7 ± 0.8 and 3.1 ± 1.4 and after intravenous infusion (both P < 0.001). After 10 and 20 mg tablets, the R-ratios were even greater. The distribution volumes at steady state of S- and R-ketamine were 6.6 ± 2.2 and 5.6 ± 2.1 l/kg, terminal half-lives 5.2 ± 3.4 and 6.1 ± 3.1 h, and metabolic clearances 1,620 ± 380 and 1,530 ± 380 ml/min, respectively. Bioavailability of the 40 mg tablets was 15 ± 8 (S-isomer) and 19 ± 10% (R-isomer) and terminal half-life 11 ± 4 and 10 ± 4 h. About 7% of the dose was renally excreted as S-stereoisomers and 17% as R-stereoisomers. CONCLUSIONS: Prolonged-release ketamine tablets generate a high systemic exposure to 2,6-hydroxynorketamines and might therefore be an efficient and safer pharmaceutical dosage form for treatment of patients with chronic neuropathic pain compared to intravenous infusion.


Asunto(s)
Analgésicos/metabolismo , Analgésicos/farmacocinética , Ketamina/metabolismo , Ketamina/farmacocinética , Administración Oral , Adulto , Analgésicos/administración & dosificación , Preparaciones de Acción Retardada , Femenino , Voluntarios Sanos , Humanos , Ketamina/administración & dosificación , Masculino , Valores de Referencia , Adulto Joven
10.
Br J Anaesth ; 127(1): 23-31, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33896589

RESUMEN

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.


Asunto(s)
Anestésicos Disociativos/química , Anestésicos Disociativos/farmacocinética , Gasto Cardíaco/efectos de los fármacos , Ketamina/química , Ketamina/farmacocinética , Adulto , Gasto Cardíaco/fisiología , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Voluntarios Sanos , Humanos , Masculino , Estereoisomerismo , Adulto Joven
11.
Eur J Clin Pharmacol ; 77(8): 1181-1192, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33575848

RESUMEN

PURPOSE: The recent repurposing of ketamine as treatment for pain and depression has increased the need for accurate population pharmacokinetic (PK) models to inform the design of new clinical trials. Therefore, the objectives of this study were to externally validate available PK models on (S)-(nor)ketamine concentrations with in-house data and to improve the best performing model when necessary. METHODS: Based on predefined criteria, five models were selected from literature. Data of two previously performed clinical trials on (S)-ketamine administration in healthy volunteers were available for validation. The predictive performances of the selected models were compared through visual predictive checks (VPCs) and calculation of the (root) mean (square) prediction errors (ME and RMSE). The available data was used to adapt the best performing model through alterations to the model structure and re-estimation of inter-individual variability (IIV). RESULTS: The model developed by Fanta et al. (Eur J Clin Pharmacol 71:441-447, 2015) performed best at predicting the (S)-ketamine concentration over time, but failed to capture the (S)-norketamine Cmax correctly. Other models with similar population demographics and study designs had estimated relatively small distribution volumes of (S)-ketamine and thus overpredicted concentrations after start of infusion, most likely due to the influence of circulatory dynamics and sampling methodology. Model predictions were improved through a reduction in complexity of the (S)-(nor)ketamine model and re-estimation of IIV. CONCLUSION: The modified model resulted in accurate predictions of both (S)-ketamine and (S)-norketamine and thereby provides a solid foundation for future simulation studies of (S)-(nor)ketamine PK in healthy volunteers after (S)-ketamine infusion.


Asunto(s)
Ketamina/análogos & derivados , Ketamina/farmacocinética , Modelos Biológicos , Adulto , Femenino , Voluntarios Sanos , Humanos , Masculino , Reproducibilidad de los Resultados , Adulto Joven
12.
Eur Arch Psychiatry Clin Neurosci ; 271(3): 417-429, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31745646

RESUMEN

The motive of this article is to review the pharmacological and clinical aspects of esketamine (ESK), an NMDA-receptor antagonist approved recently by the FDA for treatment-resistant depression (TRD). PubMed/Medline database was searched using keywords 'esketamine' and 'depression', 'S-ketamine' and 'depression', and 'NMDA antagonist' and 'depression'. Individual trials were searched from ClinicalTrials.gov. We included English-language articles evaluating pharmacokinetics and pharmacodynamics of intranasal (IN) esketamine, along with clinical trial data related to its efficacy and safety in patients diagnosed with TRD. Compared to placebo, IN esketamine causes significant and rapid improvement in depression. Dizziness, vertigo, headache, increase in blood pressure are some of its common adverse effects. With the growing number of patients of TRD, additional effective and safe treatment is the need of the hour. Esketamine appears to be an effective therapy when combined with oral antidepressants in patients with TRD. It is of special value due to the rapid onset of its action. Long-term clinical studies are, however, needed to ascertain its safety profile.


Asunto(s)
Antidepresivos/farmacología , Ensayos Clínicos Fase III como Asunto , Trastorno Depresivo Resistente al Tratamiento/tratamiento farmacológico , Antagonistas de Aminoácidos Excitadores/farmacología , Ketamina/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Quimioterapia Combinada , Antagonistas de Aminoácidos Excitadores/efectos adversos , Antagonistas de Aminoácidos Excitadores/farmacocinética , Humanos , Ketamina/efectos adversos , Ketamina/farmacocinética
13.
Eur J Clin Pharmacol ; 77(5): 671-676, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33210159

RESUMEN

PURPOSE: Ketamine has rapid-onset antidepressant effects in patients with treatment-resistant depression. Common side effects include dissociation (a sense of detachment from reality) and increases in systolic and diastolic blood pressure. The objective of this structured review was to examine the effect of ketamine formulation and route of administration on its pharmacokinetics, safety and tolerability, to identify formulation characteristics and routes of administration that might minimise side effects. METHODS: This was a structured review of published ketamine pharmacokinetics, safety and tolerability data for any ketamine formulation. The ratio of ketamine:norketamine was calculated from reported Cmax values, as a measure of first pass metabolism. The effect of formulation and route of administration on safety was evaluated by measuring mean changes in systolic blood pressure and tolerability by changes in dissociation ratings. Data were correlated using Spearman's method. RESULTS: A total of 41 treatment arms were identified from 21 publications, and included formulation development studies in healthy volunteers, and studies in clinical populations (patients undergoing anaesthesia, or being treated for pain or depression). Ketamine:norketamine ratios were strongly positively correlated with change in dissociation ratings (r = 0.89) and change in blood pressure (r = 0.96), and strongly negatively correlated with ketamine Tmax (r = - 0.87; p < 0.00001 for all). Ketamine Tmax strongly positively correlated with a change in dissociation ratings (r = - 0.96) and change in blood pressure (r = - 0.99; p < 0.00001 for all). CONCLUSION: Ketamine formulations that maximize first pass metabolism and delay Tmax will be better tolerated and safer than formulations which lack those characteristics.


Asunto(s)
Antidepresivos/administración & dosificación , Antidepresivos/efectos adversos , Sistemas de Liberación de Medicamentos/métodos , Ketamina/administración & dosificación , Ketamina/efectos adversos , Antidepresivos/farmacocinética , Trastornos Disociativos/inducido químicamente , Vías de Administración de Medicamentos , Humanos , Hipertensión/inducido químicamente , Ketamina/análogos & derivados , Ketamina/sangre , Ketamina/farmacocinética , Tasa de Depuración Metabólica
14.
Anesthesiology ; 133(6): 1192-1213, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32997732

RESUMEN

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.


Asunto(s)
Anestésicos Disociativos/farmacocinética , Ketamina/farmacocinética , Adulto , Niño , Humanos
15.
JAMA Netw Open ; 3(8): e2013211, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32785636

RESUMEN

Importance: A single subanesthetic dose of ketamine produces an antidepressant response in patients with major depressive disorder (MDD) within hours, but the mechanism of antidepressant effect is uncertain. Objective: To evaluate whether ketamine dose and brain glutamate and glutamine (Glx) and γ-aminobutyric acid (GABA) level responses to ketamine are related to antidepressant benefit and adverse effects. Design, Setting, and Participants: This randomized, parallel-group, triple-masked clinical trial included 38 physically healthy, psychotropic medication-free adult outpatients who were in a major depressive episode of MDD but not actively suicidal. The trial was conducted at Columbia University Medical Center. Data were collected from February 2012 to May 2015. Data analysis was conducted from January to March 2020. Intervention: Participants received 1 dose of placebo or ketamine (0.1, 0.2, 0.3, 0.4, or 0.5 mg/kg) intravenously during 40 minutes of a proton magnetic resonance spectroscopy scan that measured ventro-medial prefrontal cortex Glx and GABA levels in 13-minute data frames. Main Outcomes and Measures: Clinical improvement was measured using a 22-item version of the Hamilton Depression Rating Scale (HDRS-22) 24 hours after ketamine was administered. Ketamine and metabolite blood levels were measured after the scan. Results: A total of 38 individuals participated in the study, with a mean (SD) age of 38.6 (11.2) years, 23 (60.5%) women, and 25 (65.8%) White patients. Improvement in HDRS-22 score at 24 hours correlated positively with ketamine dose (t36 = 2.81; P = .008; slope estimate, 19.80 [95% CI, 5.49 to 34.11]) and blood level (t36 = 2.25; P = .03; slope estimate, 0.070 [95% CI, 0.007 to 0.133]). The lower the Glx response, the better the antidepressant response (t33 = -2.400; P = .02; slope estimate, -9.85 [95% CI, -18.2 to -1.50]). Although GABA levels correlated with Glx (t33 = 8.117; P < .001; slope estimate, 0.510 [95% CI, 0.382 to 0.638]), GABA response did not correlate with antidepressant effect. When both ketamine dose and Glx response were included in a mediation analysis model, ketamine dose was no longer associated with antidepressant effect, indicating that Glx response mediated the relationship. Adverse effects were related to blood levels in men only (t5 = 2.606; P = .048; estimated slope, 0.093 [95% CI, 0.001 to 0.186]), but Glx and GABA response were not related to adverse effects. Conclusions and Relevance: In this study, intravenous ketamine dose and blood levels correlated positively with antidepressant response. The Glx response correlated inversely with ketamine dose and with antidepressant effect. Future studies are needed to determine whether the relationship between Glx level and antidepressant effect is due to glutamate or glutamine. Trial Registration: ClinicalTrials.gov Identifier: NCT01558063.


Asunto(s)
Antidepresivos/administración & dosificación , Trastorno Depresivo Mayor , Ácido Glutámico/metabolismo , Ketamina/administración & dosificación , Ácido gamma-Aminobutírico/metabolismo , Adulto , Antidepresivos/efectos adversos , Antidepresivos/farmacocinética , Antidepresivos/uso terapéutico , Trastorno Depresivo Mayor/diagnóstico por imagen , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/metabolismo , Femenino , Humanos , Ketamina/efectos adversos , Ketamina/farmacocinética , Ketamina/uso terapéutico , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Corteza Prefrontal/diagnóstico por imagen , Corteza Prefrontal/metabolismo
16.
Br J Anaesth ; 125(5): 750-761, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32838982

RESUMEN

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.


Asunto(s)
Anestésicos Disociativos/farmacocinética , Ketamina/farmacocinética , Adulto , Anestésicos Disociativos/administración & dosificación , Biotransformación , Simulación por Computador , Estudios Cruzados , Método Doble Ciego , Composición de Medicamentos , Femenino , Humanos , Inyecciones Intravenosas , Ketamina/administración & dosificación , Ketamina/análogos & derivados , Ketamina/sangre , Ketamina/química , Masculino , Modelos Teóricos , Nitroprusiato/uso terapéutico , Complicaciones Posoperatorias/prevención & control , Complicaciones Posoperatorias/psicología , Estereoisomerismo , Adulto Joven
17.
Biomed Res Int ; 2020: 9259683, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32724819

RESUMEN

Male Sprague-Dawley rats (n = 18) were randomly divided into three groups: a saline group (20 mL/kg by gavage), a ketamine (KET) group (100 mg/kg by gavage), and a KET (the same routes and doses) combined with levo-tetrahydropalmatine (l-THP; 40 mg/kg by gavage) group (n = 6). Blood samples were acquired at different time points after drug administration. A simple and sensitive ultraperformance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method was established to determine the concentrations of KET and its metabolite, norketamine (NK), in rat plasma. Chromatographic separation was achieved using a BEH C18 column (2.1 mm × 50 mm, 1.7 µm) with chlorpheniramine maleate (Chlor-Trimeton) as an internal standard (IS). The initial mobile phase consisted of acetonitrile-water with 0.1% methanoic acid (80 : 20, v/v). The multiple reaction monitoring (MRM) modes of m/z 238.1→m/z 179.1 for KET, m/z 224.1→m/z 207.1 for NK, and m/z 275→m/z 230 for Chlor-Trimeton (IS) were utilized to conduct a quantitative analysis. Calibration curves of KET and NK in rat plasma demonstrated good linearity in the range of 2.5-500 ng/mL (r > 0.9994), and the lower limit of quantification (LLOQ) was 2.5 ng/mL for both. Moreover, the intra- and interday precision relative standard deviation (RSD) of KET and NK were less than 4.31% and 6.53%, respectively. The accuracies (relative error) of KET and NK were below -1.41% and -6.07%, respectively. The extraction recoveries of KET and NK were more than 81.23 ± 3.45% and 80.42 ± 4.57%, respectively. This sensitive, rapid, and selective UPLC-MS/MS method was successfully applied to study the pharmacokinetic effects of l-THP on KET after gastric gavage. The results demonstrated that l-THP could increase the bioavailability of KET and promote the metabolism of KET. The results showed that l-THP has pharmacokinetics effects on KET in rat plasma.


Asunto(s)
Alcaloides de Berberina/sangre , Alcaloides de Berberina/farmacocinética , Ketamina/sangre , Ketamina/farmacocinética , Plasma/química , Animales , Disponibilidad Biológica , Cromatografía Líquida de Alta Presión/métodos , Estudios de Evaluación como Asunto , Ketamina/análogos & derivados , Límite de Detección , Masculino , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem/métodos
18.
Behav Brain Res ; 390: 112631, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32437885

RESUMEN

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.


Asunto(s)
Anestésicos Disociativos/farmacología , Antidepresivos/farmacología , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/metabolismo , Ketamina/farmacología , Receptores de Neurotransmisores/efectos de los fármacos , Anestésicos Disociativos/administración & dosificación , Anestésicos Disociativos/farmacocinética , Animales , Antidepresivos/administración & dosificación , Antidepresivos/farmacocinética , Humanos , Ketamina/administración & dosificación , Ketamina/farmacocinética
19.
J Clin Psychopharmacol ; 40(3): 300-304, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32332467

RESUMEN

PURPOSE/BACKGROUND: Ketamine (K) is used as a party drug with hallucinogenic properties with a half-life of about 2.5 hours. Data are available with respect to the detection window (ie, when a person is still tested positive for the drug and/or metabolite after use) of K after single use. Nevertheless, no data are available with respect to the detection window of K in urine after chronic use. METHODS/PROCEDURES: This retrospective case series describes 7 patients with K dependency as their main addiction who have been admitted to an addiction center for K detoxification. Their abstinence-oriented care involved routine urinary screening of K and its metabolites, as well as traditional drugs of abuse, such as cocaine and cannabinoids. FINDINGS/RESULTS: Urine samples remained positive for all the cases identified after 22 to 96 days. A peak detection period of 61, 40, and 96 days for K, norketamine, and dehydronorketamine, respectively, measured using liquid chromatography-tandem mass spectrometry at a cutoff concentration of 1.0 ng/mL, is defined. The K/norketamine and K/dehydronorketamine ratios varied over time between 0.33 and 3.06, and 0.01 and 0.36 for all patients, respectively, implying a large interindividual variation in K metabolism. IMPLICATIONS/CONCLUSIONS: Ketamine and its metabolites have a prolonged excretion profile in urine, which requires frequent measurements (at least weekly) to guide abstinence treatment. Further research is needed to develop an algorithm that can differentiate new K use from residual urinary K excretion in urine of chronic daily users.


Asunto(s)
Ketamina/análogos & derivados , Ketamina/administración & dosificación , Ketamina/orina , Detección de Abuso de Sustancias/métodos , Adulto , Femenino , Humanos , Ketamina/farmacocinética , Masculino , Estudios Retrospectivos , Factores de Tiempo , Adulto Joven
20.
Vet Anaesth Analg ; 47(3): 334-340, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32222343

RESUMEN

OBJECTIVE: To describe the pharmacokinetics of ketamine following a short intravenous (IV) infusion to isoflurane-anesthetized rabbits. STUDY DESIGN: Prospective experimental study. ANIMALS: A total of six adult healthy female New Zealand White rabbits. METHODS: Anesthesia was induced with isoflurane in oxygen. Following determination of isoflurane minimum alveolar concentration (MAC), the isoflurane concentration was reduced to 0.75 MAC and ketamine hydrochloride (5 mg kg-1) was administered IV over 5 minutes. Blood samples were collected before and at 2, 5, 6, 7, 8, 9, 13, 17, 21, 35, 65, 125, 215 and 305 minutes after initiating the ketamine infusion. Samples were processed immediately and the plasma separated and stored at -80 °C until analyzed for ketamine and norketamine concentrations using liquid chromatography-mass spectrometry. Compartment models were fitted to the concentration-time data for ketamine and for ketamine plus norketamine using nonlinear mixed-effects (population) modeling. RESULTS: A three- and five-compartment model best fitted the plasma concentration-time data for ketamine and for ketamine plus norketamine, respectively. For the ketamine only model, the volume of distribution at steady state (Vss) was 3217 mL kg-1, metabolic clearance was 88 mL minute-1 kg-1 and the terminal half-life was 59 minutes. For the model including both ketamine and norketamine, Vss were 3224 and 2073 mL kg-1, total metabolic clearance was 107 and 52 mL minute-1 kg-1 and terminal half-lives were 52 and 55 minutes for the parent drug and its metabolite, respectively. CONCLUSIONS AND CLINICAL RELEVANCE: This study characterized the pharmacokinetics of ketamine and norketamine in isoflurane-anesthetized New Zealand White rabbits following short IV infusion. The results obtained herein will be useful to determine ketamine infusion regimens in isoflurane-anesthetized rabbits.


Asunto(s)
Analgésicos/farmacocinética , Anestésicos por Inhalación/administración & dosificación , Isoflurano/administración & dosificación , Ketamina/análogos & derivados , Ketamina/farmacocinética , Conejos/metabolismo , Analgésicos/administración & dosificación , Animales , Femenino , Infusiones Intravenosas/veterinaria , Ketamina/administración & dosificación , Estudios Prospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...