1
|
Guan Y, Li B, Zhang Y, Luo H, Wang X, Bai X, Zheng Z, Huang Y, Wei W, Huang M, Song X, Zhong G. Pharmacogenetic and pharmacokinetic factors for dexmedetomidine-associated hemodynamic instability in pediatric patients. Front Pharmacol 2025; 15:1515523. [PMID: 39840108 PMCID: PMC11745869 DOI: 10.3389/fphar.2024.1515523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 11/25/2024] [Indexed: 01/23/2025] Open
Abstract
Purpose The incidence of hemodynamic instability associated with dexmedetomidine (DEX) sedation has been reported to exceed 50%, with substantial inter-individual variability in response. Genetic factors have been suggested to contribute significantly to such variation. The aim of this study was to identify the clinical, pharmacokinetic, and genetic factors associated with DEX-induced hemodynamic instability in pediatric anesthesia patients. Methods A cohort of 270 pediatric patients scheduled for elective interventional surgery received an intranasal dose of 3 mcg·kg-1 of dexmedetomidine, and subsequent propofol induction was conducted when patients had a UMSS of 2-4. The primary endpoint was hemodynamic instability-defined as a composite of hypotension and/or bradycardia, which is characterized by a 20% reduction from age-specific baseline values. Plasma concentrations of dexmedetomidine were determined, and single-nucleotide polymorphisms (SNPs) were genotyped. A validated population pharmacokinetic model was used to estimate pharmacokinetic parameters. LASSO regression was used to identify significant factors, and a Cox's proportional hazards model-derived nomogram for hemodynamic instability was developed. Results Hemodynamic instability was observed in 52 out of 270 patients (209 events), resulting in a cumulative incidence of 16.30% at 90 min, as estimated by Kaplan-Meier estimation, and it was associated with a median time to event of 35 min. The interval time between DEX initiation and propofol induction was 16 min (IQR: 12-22 min). The cumulative incidence was 8.2% within 22 min after DEX initiation. The identified significant risk factors for DEX-associated hemodynamic instability included weight, DEX clearance, concomitant propofol use, and the following gene variants UGT2B10 rs1841042 (hazard ratio (HR):1.41, 95% confidence interval (CI): 1.12-1.79), CYP2A6 rs8192733 (HR:0.28, 95%CI:0.09-0.88), ADRA2B rs3813662 (HR:1.39,95%CI:1.02-1.89), CACNA2D2 rs2236957 (HR:1.46, 95%CI:1.09-1.96), NR1I2 rs3814057 (HR:0.64, 95%CI:0.43-0.95), and CACNB2 rs10764319 (HR:1.40,95%CI:1.05-1.87). The areas under the curve for the training and test cohorts were 0.881 and 0.762, respectively. The calibration curve indicated excellent agreement. Conclusion The predictive nomogram, which incorporates genetic variants (UGT2B10, CYP2A6, ADRA2B, CACNA2D2, NR1I2, and CACNB2) along with clinical factors such as weight, DEX clearance, and propofol use, may help prevent DEX-associated hemodynamic instability. Delayed hemodynamic instability is likely to occur after 35-min DEX initiation in patients with lower DEX clearance after propofol induction.
Collapse
Affiliation(s)
- Yanping Guan
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Bilian Li
- Department of Anesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yiyu Zhang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Hao Luo
- Department of Anesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Xueding Wang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Xue Bai
- Department of Anesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Zhuoling Zheng
- Department of Pharmacy, Sun Yat-sen University Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yaying Huang
- Department of Anesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Wei Wei
- Department of Anesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Min Huang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Xingrong Song
- Department of Anesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Guoping Zhong
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
Vaz Rodrigues L, Roriz D, Salgado-Seixas F, Marinho S, Ferreira PR. A nephrogenic mechanism underlies dexmedetomidine-induced polyuria. A case report. REVISTA ESPANOLA DE ANESTESIOLOGIA Y REANIMACION 2024; 71:332-338. [PMID: 37717633 DOI: 10.1016/j.redare.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/28/2022] [Indexed: 09/19/2023]
Abstract
Dexmedetomidine's α-adrenoreceptor agonism has been gaining popularity in the anesthetic room as a sedative-hypnotic and analgesic agent, and with extensive perioperative use rising concern about side effects is necessary. Bradycardia and hypotension are common adverse effects, but there are also several reports of excessive urine output, possibly due to vasopressin secretion and permeability of collecting ducts. Polyuria usually resolves with discontinuation of the drug, and significant morbidity has not been reported. Early identification, removal of the agent, and treatment are imperative to minimize complications - mainly natremia and neurological symptoms. This case report describes a dexmedetomidine-related polyuric syndrome during opioid-free general anesthesia for major head and neck surgery. A nephrogenic mechanism for the clinical effect is proposed and reinforced by analytical data obtained. An intra-operative polyuria approach is also delineated.
Collapse
Affiliation(s)
| | - D Roriz
- Centro Hospitalar de Trás-os-Montes e Alto Douro E.P.E., Vila Real, Portugal
| | | | - S Marinho
- Hospital do Divino Espírito Santo de Ponta Delgada, Ponta Delgada, Portugal
| | - P R Ferreira
- Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| |
Collapse
|
3
|
Dzierba AL, Stollings JL, Devlin JW. A pharmacogenetic precision medicine approach to analgesia and sedation optimization in critically ill adults. Pharmacotherapy 2023; 43:1154-1165. [PMID: 36680385 DOI: 10.1002/phar.2768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 01/22/2023]
Abstract
Precision medicine is a growing field in critical care. Research increasingly demonstrated pharmacogenomic variability to be an important determinant of analgesic and sedative drug response in the intensive care unit (ICU). Genome-wide association and candidate gene finding studies suggest analgesic and sedatives tailored to an individual's genetic makeup, environmental adaptations, in addition to several other patient- and drug-related factors, will maximize effectiveness and help mitigate harm. However, the number of pharmacogenetic studies in ICU patients remains small and no prospective studies have been published using pharmacogenomic data to optimize analgesic or sedative therapy in critically ill patients. Current recommendations for treating ICU pain and agitation are based on controlled studies having low external validity, including the failure to consider pharmacogenomic factors affecting response. Use of a precision medicine approach to individualize pharmacotherapy focused on optimizing ICU patient comfort and safety may improve the outcomes of critically ill adults. Additionally, benefits and risks of analgesic and/or sedative therapy in an individual may be informed with large, standardized datasets. The purpose of this review was to describe a precision medicine approach focused on optimizing analgesic and sedative therapy in individual ICU patients to optimize clinical outcomes and reduce safety concerns.
Collapse
Affiliation(s)
- Amy L Dzierba
- Department of Pharmacy, New York-Presbyterian Hospital, New York, New York, USA
- Center for Acute Respiratory Failure, Columbia University College of Physicians and Surgeons and New York-Presbyterian Hospital, New York, New York, USA
| | - Joanna L Stollings
- Department of Pharmaceutical Services, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Nashville, Tennessee, USA
| | - John W Devlin
- School of Pharmacy, Northeastern University, Boston, Massachusetts, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Shannon ML, Muhammad A, James NT, Williams ML, Breeyear J, Edwards T, Mosley JD, Choi L, Kannankeril P, Van Driest S. Variant-based heritability assessment of dexmedetomidine and fentanyl clearance in pediatric patients. Clin Transl Sci 2023; 16:1628-1638. [PMID: 37353859 PMCID: PMC10499425 DOI: 10.1111/cts.13574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/12/2023] [Accepted: 06/01/2023] [Indexed: 06/25/2023] Open
Abstract
Despite complex pathways of drug disposition, clinical pharmacogenetic predictors currently rely on only a few high effect variants. Quantification of the polygenic contribution to variability in drug disposition is necessary to prioritize target drugs for pharmacogenomic approaches and guide analytic methods. Dexmedetomidine and fentanyl, often used in postoperative care of pediatric patients, have high rates of inter-individual variability in dosing requirements. Analyzing previously generated population pharmacokinetic parameters, we used Bayesian hierarchical mixed modeling to measure narrow-sense (additive) heritability (h SNP 2 ) of dexmedetomidine and fentanyl clearance in children and identify relative contributions of small, moderate, and large effect-size variants toh SNP 2 . We used genome-wide association studies (GWAS) to identify variants contributing to variation in dexmedetomidine and fentanyl clearance, followed by functional analyses to identify associated pathways. For dexmedetomidine, median clearance was 33.0 L/h (interquartile range [IQR] 23.8-47.9 L/h) andh SNP 2 was estimated to be 0.35 (90% credible interval 0.00-0.90), with 45% ofh SNP 2 attributed to large-, 32% to moderate-, and 23% to small-effect variants. The fentanyl cohort had median clearance of 8.2 L/h (IQR 4.7-16.7 L/h), with estimatedh SNP 2 of 0.30 (90% credible interval 0.00-0.84). Large-effect variants accounted for 30% ofh SNP 2 , whereas moderate- and small-effect variants accounted for 37% and 33%, respectively. As expected, given small sample sizes, no individual variants or pathways were significantly associated with dexmedetomidine or fentanyl clearance by GWAS. We conclude that clearance of both drugs is highly polygenic, motivating the future use of polygenic risk scores to guide appropriate dosing of dexmedetomidine and fentanyl.
Collapse
Affiliation(s)
| | - Ayesha Muhammad
- School of MedicineVanderbilt UniversityNashvilleTennesseeUSA
| | - Nathan T. James
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
- Present address:
Berry Consultants, LLCAustinTexasUSA
| | - Michael L. Williams
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
- Present address:
Department of Clinical Pharmacology and Quantitative PharmacologyAstraZenecaGothenburgSweden
| | - Joseph Breeyear
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Todd Edwards
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jonathan D. Mosley
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Leena Choi
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Prince Kannankeril
- Center for Pediatric Precision Medicine, Department of PediatricsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Sara Van Driest
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Center for Pediatric Precision Medicine, Department of PediatricsVanderbilt University Medical CenterNashvilleTennesseeUSA
- Present address:
All of Us Research ProgramNational Institutes of HealthWashingtonDCUSA
| |
Collapse
|
5
|
Jones JD, Rajachandran L, Yocca F, Risinger R, De Vivo M, Sabados J, Levin FR, Comer SD. Sublingual dexmedetomidine (BXCL501) reduces opioid withdrawal symptoms: findings from a multi-site, phase 1b/2, randomized, double-blind, placebo-controlled trial. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2023; 49:109-122. [PMID: 36630319 PMCID: PMC11036405 DOI: 10.1080/00952990.2022.2144743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/30/2022] [Accepted: 11/03/2022] [Indexed: 01/12/2023]
Abstract
Background: Like other alpha-2-adrenergic receptor agonists, dexmedetomidine may reduce the severity of opioid withdrawal but with fewer adverse cardiovascular effects.Objective: This study assessed the safety of sublingual dexmedetomidine (BXCL501) and its preliminary efficacy in treating opioid withdrawal (ClinicalTrials.gov: NCT04470050).Methods: Withdrawal was induced among individuals with physiological dependence on opioids via discontinuation of oral morphine (Days 1-5). Participants were randomized to receive placebo or active BXCL501: 30, 60, 90, 120, 180, and 240 μg twice daily (Days 6-12). Treatment-emergent adverse events (TEAEs) were the primary outcome measure. Secondary outcomes included the Clinical and Subjective Opiate Withdrawal Scales (COWS and SOWS-Gossop, respectively), and the Agitation and Calmness Evaluation Scale (ACES).Results: Of 225 participants enrolled, 90 discontinued during morphine stabilization. Post-BXCL501 randomization (Day 6) data were available from 135 participants (73% male), with 33% completing thru Day 12. In total, 36 subjects reported 1 or more TEAE. Higher doses of BXCL501 (i.e. 180 and 240 µg, twice daily) increased the frequency of: hypotension, orthostatic hypotension, and somnolence. TEAEs related to BXCL501 were mild or moderate in severity, except for one participant in the 120 µg condition whose orthostatic hypotension and bradycardia were classified as severe. Higher BXCL501 dose conditions (120, 180, and 240 µg) resulted in statistically significant reductions in COWS & SOWS scores. Mean ratings on the ACES were between 3 (mild), 4 (normal), and 5 (mild calmness), with few significant differences as a function of dose.Conclusions: These findings support the continued development of BXCL501 for the management of opioid withdrawal.
Collapse
Affiliation(s)
- Jermaine D. Jones
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute and Columbia University Irving Medical Center, New York, NY, USA
| | | | - Frank Yocca
- BioXcel Therapeutics, Inc, New Haven, CT, USA
| | | | | | | | - Frances R. Levin
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute and Columbia University Irving Medical Center, New York, NY, USA
| | - Sandra D. Comer
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute and Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
6
|
Fang C, Ouyang W, Zeng Y, Pei Q, Xia Y, Luo S, Chen M. CYP2A6 and GABRA2 Gene Polymorphisms are Associated With Dexmedetomidine Drug Response. Front Pharmacol 2022; 13:943200. [PMID: 35873555 PMCID: PMC9301121 DOI: 10.3389/fphar.2022.943200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/20/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Dexmedetomidine is a commonly used clinical sedative; however, the drug response varies among individuals. Thus, the purpose of this study was to explore the association between dexmedetomidine response and gene polymorphisms related to drug-metabolizing enzymes and drug response (CYP2A6, UGT2B10, UGT1A4, ADRA2A, ADRA2B, ADRA2C, GABRA1, GABRB2, and GLRA1). Methods: This study was a prospective cohort study. A total of 194 female patients aged 18–60 years, American Society of Anesthesiologists (ASA) score I-II, who underwent laparoscopy at the Third Xiangya Hospital of Central South University, were included. The sedative effect was assessed every 2 min using the Ramsay score, and the patient’s heart rate decrease within 20 min was recorded. Peripheral blood was collected from each participant to identify genetic variants in the candidate genes of metabolic and drug effects using the Sequenom MassARRAY® platform. Furthermore, additional peripheral blood samples were collected from the first 99 participants at multiple time points after dexmedetomidine infusion to perform dexmedetomidine pharmacokinetic analysis by Phoenix® WinNonlin 7.0 software. Results: Carriers of the minor allele (C) of CYP2A6 rs28399433 had lower metabolic enzyme efficiency and higher plasma concentrations of dexmedetomidine. In addition, the participants were divided into dexmedetomidine sensitive or dexmedetomidine tolerant groups based on whether they had a Ramsay score of at least four within 20 min, and CYP2A6 rs28399433 was identified to have a significant influence on the dexmedetomidine sedation sensitivity by logistic regression with Plink software [p = 0.003, OR (95% CI): 0.27 (0.11–0.65)]. C allele carriers were more sensitive to the sedative effects of dexmedetomidine than A allele carriers. GABRA2 rs279847 polymorphism was significantly associated with the degree of the heart rate decrease. In particular, individuals with the GG genotype had a 4-fold higher risk of heart rate abnormality than carriers of the T allele (OR = 4.32, 95% CI: 1.96–9.50, p = 0.00027). Conclusion:CYP2A6 rs28399433 polymorphism affects the metabolic rate of dexmedetomidine and is associated with susceptibility to the sedative effects of dexmedetomidine; GABRA2 rs279847 polymorphism is significantly associated with the degree of the heart rate decrease.
Collapse
Affiliation(s)
- Chao Fang
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wen Ouyang
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Youjie Zeng
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Qi Pei
- Department of Pharmacy, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuhao Xia
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Siwan Luo
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Minghua Chen
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Minghua Chen,
| |
Collapse
|
7
|
James NT, Breeyear JH, Caprioli R, Edwards T, Hachey B, Kannankeril PJ, Keaton JM, Marshall MD, Van Driest SL, Choi L. Population pharmacokinetic analysis of dexmedetomidine in children using real-world data from electronic health records and remnant specimens. Br J Clin Pharmacol 2022; 88:2885-2898. [PMID: 34957589 PMCID: PMC9106818 DOI: 10.1111/bcp.15194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 11/18/2021] [Accepted: 12/14/2021] [Indexed: 11/30/2022] Open
Abstract
AIMS Our objectives were to perform a population pharmacokinetic analysis of dexmedetomidine in children using remnant specimens and electronic health records (EHRs) and explore the impact of patient's characteristics and pharmacogenetics on dexmedetomidine clearance. METHODS Dexmedetomidine dosing and patient data were gathered from EHRs and combined with opportunistically sampled remnant specimens. Population pharmacokinetic models were developed using nonlinear mixed-effects modelling. Stage 1 developed a model without genotype variables; Stage 2 added pharmacogenetic effects. RESULTS Our final study population included 354 post-cardiac surgery patients aged 0-22 years (median 16 mo). The data were best described with a 2-compartment model with allometric scaling for weight and Hill maturation function for age. Population parameter estimates and 95% confidence intervals were 27.3 L/h (24.0-31.1 L/h) for total clearance, 161 L (139-187 L) for central compartment volume of distribution, 26.0 L/h (22.5-30.0 L/h) for intercompartmental clearance and 7903 L (5617-11 119 L) for peripheral compartment volume of distribution. The estimate for postmenstrual age when 50% of adult clearance is achieved was 42.0 weeks (41.5-42.5 weeks) and the Hill coefficient estimate was 7.04 (6.99-7.08). Genotype was not statistically or clinically significant. CONCLUSION Our study demonstrates the use of real-world EHR data and remnant specimens to perform a population pharmacokinetic analysis and investigate covariate effects in a large paediatric population. Weight and age were important predictors of clearance. We did not find evidence for pharmacogenetic effects of UGT1A4 or UGT2B10 genotype or CYP2A6 risk score.
Collapse
Affiliation(s)
- Nathan T. James
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Joseph H. Breeyear
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Richard Caprioli
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Todd Edwards
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Brian Hachey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Prince J. Kannankeril
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
- Center for Pediatric Precision Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Jacob M. Keaton
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Matthew D. Marshall
- Department of Pharmaceutical Services, Vanderbilt University Medical Center, Nashville, TN
| | - Sara L. Van Driest
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
- Center for Pediatric Precision Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Leena Choi
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
8
|
Dexmedetomidine Clearance Decreases with Increasing Drug Exposure: Implications for Current Dosing Regimens and Target-controlled Infusion Models Assuming Linear Pharmacokinetics. Anesthesiology 2021; 136:279-292. [PMID: 34851425 DOI: 10.1097/aln.0000000000004049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Numerous pharmacokinetic models have been published aiming at more accurate and safer dosing of dexmedetomidine. The vast majority of the developed models underpredict the measured plasma concentrations with respect to the target concentration, especially at plasma concentrations higher than those used in the original studies. The aim of this article was to develop a dexmedetomidine pharmacokinetic model in healthy adults emphasizing linear versus nonlinear kinetics. METHODS The data of two previously published clinical trials with stepwise increasing dexmedetomidine target-controlled infusion were pooled to build a pharmacokinetic model using the NONMEM software package (ICON Development Solutions, USA). Data from 48 healthy subjects, included in a stratified manner, were utilized to build the model. RESULTS A three-compartment mamillary model with nonlinear elimination from the central compartment was superior to a model assuming linear pharmacokinetics. Covariates included in the final model were age, sex, and total body weight. Cardiac output did not explain between-subject or within-subject variability in dexmedetomidine clearance. The results of a simulation study based on the final model showed that at concentrations up to 2 ng · ml-1, the predicted dexmedetomidine plasma concentrations were similar between the currently available Hannivoort model assuming linear pharmacokinetics and the nonlinear model developed in this study. At higher simulated plasma concentrations, exposure increased nonlinearly with target concentration due to the decreasing dexmedetomidine clearance with increasing plasma concentrations. Simulations also show that currently approved dosing regimens in the intensive care unit may potentially lead to higher-than-expected dexmedetomidine plasma concentrations. CONCLUSIONS This study developed a nonlinear three-compartment pharmacokinetic model that accurately described dexmedetomidine plasma concentrations. Dexmedetomidine may be safely administered up to target-controlled infusion targets under 2 ng · ml-1 using the Hannivoort model, which assumed linear pharmacokinetics. Consideration should be taken during long-term administration and during an initial loading dose when following the dosing strategies of the current guidelines. EDITOR’S PERSPECTIVE
Collapse
|
9
|
Packiasabapathy S, Rangasamy V, Horn N, Hendrickson M, Renschler J, Sadhasivam S. Personalized pediatric anesthesia and pain management: problem-based review. Pharmacogenomics 2020; 21:55-73. [PMID: 31849281 DOI: 10.2217/pgs-2019-0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pharmacogenetics, the genetic influence on the interpersonal variability in drug response, has enabled tailored pharmacotherapy and emerging 'personalized medicine.' Although oncology spearheaded the clinical implementation of personalized medicine, other specialties are rapidly catching up. In anesthesia, classical examples of genetically mediated idiosyncratic reactions have been long known (e.g., malignant hyperthermia and prolonged apnea after succinylcholine). The last two decades have witnessed an expanding body of pharmacogenetic evidence in anesthesia. This review highlights some of the prominent pharmacogenetic associations studied in anesthesia and pain management, with special focus on pediatric anesthesia.
Collapse
Affiliation(s)
- Senthil Packiasabapathy
- Department of Anesthesia, Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, IN 46202, USA
| | - Valluvan Rangasamy
- Department of Anesthesia, Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, IN 46202, USA
| | - Nicole Horn
- Department of Anesthesia, Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, IN 46202, USA
| | - Michele Hendrickson
- Department of Anesthesia, Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, IN 46202, USA
| | - Janelle Renschler
- Department of Anesthesia, Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, IN 46202, USA
| | - Senthilkumar Sadhasivam
- Department of Anesthesia, Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, IN 46202, USA
| |
Collapse
|
10
|
Effects of Dexmedetomidine on the Pharmacokinetics of Parecoxib and Its Metabolite Valdecoxib in Beagles by UPLC-MS/MS. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1563874. [PMID: 32832543 PMCID: PMC7428950 DOI: 10.1155/2020/1563874] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/06/2020] [Accepted: 07/07/2020] [Indexed: 11/29/2022]
Abstract
A sensitive and reliable ultraperformance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method was developed for the simultaneous determination of parecoxib and its metabolite valdecoxib in beagles. The effects of dexmedetomidine on the pharmacokinetics of parecoxib and valdecoxib in beagles were studied. The plasma was precipitated by acetonitrile, and the two analytes were separated on an Acquity UPLC BEH C18 column (2.1 mm × 50 mm, 1.7 μm); the mobile phase was acetonitrile and 0.1% formic acid with gradient mode, and the flow rate was 0.4 mL/min. In the negative ion mode, the two analytes and internal standard (IS) were monitored by multiple reaction monitoring (MRM), and the mass transition pairs were as follows: m/z 369.1 → 119.1 for parecoxib, m/z 313.0 → 118.0 for valdecoxib, and m/z 380.0 → 316.0 for celecoxib (IS). Six beagles were designed as a double cycle self-control experiment. In the first cycle, after intramuscular injection of parecoxib 1.33 mg/kg, 1.0 mL blood samples were collected at different times (group A). In the second cycle, the same six beagles were intravenously injected with 2 μg/kg dexmedetomidine for 7 days after one week of washing period. On day 7, after intravenous injection of 2 μg/kg dexmedetomidine for 0.5 hours, 6 beagle dogs were intramuscularly injected with 1.33 mg/kg parecoxib, and blood samples were collected at different time points (group A). The concentration of parecoxib and valdecoxib was detected by UPLC-MS/MS, and the main pharmacokinetic parameters were calculated by DAS 2.0 software. Under the experimental conditions, the method has a good linear relationship for both analytes. The interday and intraday precision was less than 8.07%; the accuracy values were from -1.20% to 2.76%. Cmax of parecoxib in group A and group B was 2148.59 ± 406.13 ng/mL and 2100.49 ± 356.94 ng/mL, t1/2 was 0.85 ± 0.36 h and 0.85 ± 0.36 h, and AUC(0‐t) was 2429.96 ± 323.22 ng·h/mL and 2506.38 ± 544.83 ng·h/mL, respectively. Cmax of valdecoxib in group A and group B was 2059.15 ± 281.86 ng/mL and 2837.39 ± 276.78 ng/mL, t1/2 was 2.44 ± 1.55 h and 2.91 ± 1.27 h, and AUC(0‐t) was 4971.61 ± 696.56 ng·h/mL and 6770.65 ± 453.25 ng·h/mL, respectively. There was no significant change in the pharmacokinetics of parecoxib in groups A and B. Cmax and AUC(0 − ∞) of valdecoxib in group A were 37.79% and 36.19% higher than those in group B, respectively, and t1/2 was increased from 2.44 h to 2.91 h. Vz/F and CLz/F were correspondingly reduced, respectively. The developed UPLC-MS/MS method for simultaneous determination of parecoxib and valdecoxib in beagle plasma was specific, accurate, rapid, and suitable for the pharmacokinetics and drug-drug interactions of parecoxib and valdecoxib. Dexmedetomidine can inhibit the metabolism of valdecoxib in beagles and increase the exposure of valdecoxib, but it does not affect the pharmacokinetics of parecoxib.
Collapse
|
11
|
Zhou W, Li SL, Zhao T, Li L, Xing WB, Qiu XJ, Zhang W. Effects of Dexmedetomidine on the Pharmacokinetics of Dezocine, Midazolam and Its Metabolite 1-Hydroxymidazolam in Beagles by UPLC-MS/MS. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2595-2605. [PMID: 32753841 PMCID: PMC7342499 DOI: 10.2147/dddt.s254055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/12/2020] [Indexed: 01/13/2023]
Abstract
Objective We developed and validated a sensitive and reliable UPLC-MS/MS method for simultaneous determination of dezocine (DEZ), midazolam (MDZ) and its metabolite 1-hydroxymidazolam (1-OH-MDZ) in beagle plasma and investigated the effect of dexmedetomidine (DEX) on the pharmacokinetics of DEZ, MDZ and 1-OH-MDZ in beagles. Materials and Methods Diazepam was used as the internal standard (IS); the three analytes and IS were extracted by acetonitrile precipitation and separated on an Acquity UPLC BEH C18 column using acetonitrile-0.1% formic acid as mobile phase in gradient mode. In positive ion mode, the three analytes and IS were monitored by multiple reaction monitoring (MRM). Six beagles were designed as a double cycle self-control experiment with 0.15 mg/kg in the first cycle (Group A). After a 1-week washout period, the same six beagles were slowly injected intravenously with 2 µg/kg DEX in the second cycle (Group B), with continuous injection for 7 days. On the seventh day, 0.5 hr after intravenous injection of 2 µg/kg DEX, the six beagles were intramuscularly given with DEZ 0.33 mg/kg and MDZ 0.15 mg/kg. Results Under the conditions of this experiment, this method exhibited a good linearity for each analyte. The accuracy and precision were all within the acceptable limits in the bioanalytical method, and the results of recovery, matrix effect and stability have also met the requirements. Conclusion The developed UPLC-MS/MS method for simultaneous determination of DEZ, MDZ and 1-OH-MDZ in beagles plasma was accurate, reproducible, specific, and suitable. DEX could inhibit the metabolism of DEZ and MDZ and increase the exposure of DEZ and MDZ in beagles. Therefore, the change of therapeutic effect and the occurrence of adverse reactions caused by drug–drug interaction should be paid attention to when the drugs were used in combination.
Collapse
Affiliation(s)
- Wei Zhou
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China.,Nanyang City Central Hospital, Nanyang 473009, People's Republic of China
| | - Shuang-Long Li
- School of Basic Medicine, Henan University of Science and Technology, Luoyang 471023, People's Republic of China
| | - Ti Zhao
- School of Basic Medicine, Henan University of Science and Technology, Luoyang 471023, People's Republic of China
| | - Le Li
- School of Basic Medicine, Henan University of Science and Technology, Luoyang 471023, People's Republic of China
| | - Wen-Bin Xing
- School of Basic Medicine, Henan University of Science and Technology, Luoyang 471023, People's Republic of China
| | - Xiang-Jun Qiu
- School of Basic Medicine, Henan University of Science and Technology, Luoyang 471023, People's Republic of China
| | - Wei Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| |
Collapse
|
12
|
Ber J, Wiczling P, Hołysz M, Klupczyńska A, Bartkowska-Śniatkowska A, Bieda K, Smuszkiewicz P, Nowicka M, Żurański Ł, Sobczyński P, Matysiak J, Grześkowiak E, Bienert A. Population Pharmacokinetic Model of Dexmedetomidine in a Heterogeneous Group of Patients. J Clin Pharmacol 2020; 60:1461-1473. [PMID: 32500578 DOI: 10.1002/jcph.1647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 04/28/2020] [Indexed: 11/11/2022]
Abstract
Dexmedetomidine is a hepatically eliminated drug with sedative, anxiolytic, sympatholytic, and analgesic properties that has been increasingly used for various indications in the form of a short or continuous intravenous infusion. This study aimed to propose a population pharmacokinetic (PK) model of dexmedetomidine in a heterogeneous group of intensive care unit patients, incorporating 29 covariates potentially linked with dexmedetomidine PK. Data were collected from 70 patients aged between 0.25 and 88 years and treated with dexmedetomidine infusion for various durations at 1 of 4 medical centers. Statistical analysis was performed using a nonlinear mixed-effect model. Categorical and continuous covariates including demographic data, hemodynamic parameters, biochemical markers, and 11 single-nucleotide polymorphisms were tested. A 2-compartment model was used to describe dexmedetomidine PK. An allometric/isometric scaling was used to account for body weight difference in PK parameters, and the Hill equation was used to describe the maturation of clearance. Typical values of the central and peripheral volume of distribution and the systemic and distribution clearance for a theoretical adult patient were central volume of distribution = 22.50 L, peripheral volume of distribution = 86.1 L, systemic clearance = 34.7 L/h, and distribution clearance = 40.8 L/h. The CYP1A2 genetic polymorphism and noradrenaline administration were identified as significant covariates for clearance. A population PK model of dexmedetomidine was successfully developed. The proposed model is well calibrated to the observed data. The identified covariates account for <5% of interindividual variability and consequently are of low clinical significance for the purpose of dose adjustment.
Collapse
Affiliation(s)
- Justyna Ber
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, Poznan, Poland
| | - Paweł Wiczling
- Department of Biopharmacy and Pharmacodynamics, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Hołysz
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, Poznan, Poland
| | - Agnieszka Klupczyńska
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Krzysztof Bieda
- Anaesthesiology and Intensive Care Department, Greater Poland Cancer Centre, Poznan, Poland
| | - Piotr Smuszkiewicz
- Department of Anesthesiology, Intensive Therapy and Pain Treatment, Heliodor Swiecicki Clinical Hospital, Poznan University of Medical Sciences, Poznan, Poland
| | - Małgorzata Nowicka
- Anaesthetics and Critical Care Department, University Hospital of Lord's Transfiguration, Poznan University of Medical Sciences, Poznan, Poland
| | - Łukasz Żurański
- Anaesthetics and Critical Care Department, University Hospital of Lord's Transfiguration, Poznan University of Medical Sciences, Poznan, Poland
| | - Paweł Sobczyński
- Anaesthetics and Critical Care Department, University Hospital of Lord's Transfiguration, Poznan University of Medical Sciences, Poznan, Poland
| | - Jan Matysiak
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Poznan, Poland
| | - Edmund Grześkowiak
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, Poznan, Poland
| | - Agnieszka Bienert
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
13
|
Anderson BJ, Morse JD, Hannam JA, Cortinez LI. Pharmacokinetic and pharmacodynamic considerations of general anesthesia in pediatric subjects. Expert Opin Drug Metab Toxicol 2020; 16:279-295. [PMID: 32148110 DOI: 10.1080/17425255.2020.1739648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Introduction: The target concentration strategy uses PKPD information for dose determination. Models have also quantified exposure-response relationships, improved understanding of developmental pharmacokinetics, rationalized dose prescription, provided insight into the importance of covariate information, explained drug interactions and driven decision-making and learning during drug development.Areas covered: The prime PKPD consideration is parameter estimation and quantification of variability. The main sources of variability in children are age (maturation) and weight (size). Model use is mostly confined to pharmacokinetics, partly because anesthesia effect models in the young are imprecise. Exploration of PK and PD covariates and their variability hold potential to better individualize treatment.Expert opinion: The ability to model drugs using computer-based technology is hindered because covariate data required to individualize treatment using these programs remain lacking. Target concentration intervention strategies remain incomplete because covariate information that might better predict individualization of dose is absent. Pharmacogenomics appear a valuable area for investigation for pharmacodynamics and pharmacodynamics. Effect measures in the very young are imprecise. Assessment of the analgesic component of anesthesia is crude. While neuromuscular monitoring is satisfactory, depth of anaesthesia EEG interpretation is inadequate. Closed loop anesthesia is possible with better understanding of EEG changes.
Collapse
Affiliation(s)
- Brian J Anderson
- Department of Anaesthesiology, University of Auckland, Auckland, New Zealand
| | - James D Morse
- Department of Pharmacology & Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Jacqueline A Hannam
- Department of Pharmacology & Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - L Ignacio Cortinez
- División Anestesiología, Pontificia Universidad Católica De Chile, Santiago De Chile, Chile
| |
Collapse
|
14
|
Castillo RL, Ibacache M, Cortínez I, Carrasco-Pozo C, Farías JG, Carrasco RA, Vargas-Errázuriz P, Ramos D, Benavente R, Torres DH, Méndez A. Dexmedetomidine Improves Cardiovascular and Ventilatory Outcomes in Critically Ill Patients: Basic and Clinical Approaches. Front Pharmacol 2020; 10:1641. [PMID: 32184718 PMCID: PMC7058802 DOI: 10.3389/fphar.2019.01641] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Dexmedetomidine (DEX) is a highly selective α2-adrenergic agonist with sedative and analgesic properties, with minimal respiratory effects. It is used as a sedative in the intensive care unit and the operating room. The opioid-sparing effect and the absence of respiratory effects make dexmedetomidine an attractive adjuvant drug for anesthesia in obese patients who are at an increased risk for postoperative respiratory complications. The pharmacodynamic effects on the cardiovascular system are known; however the mechanisms that induce cardioprotection are still under study. Regarding the pharmacokinetics properties, this drug is extensively metabolized in the liver by the uridine diphosphate glucuronosyltransferases. It has a relatively high hepatic extraction ratio, and therefore, its metabolism is dependent on liver blood flow. This review shows, from a basic clinical approach, the evidence supporting the use of dexmedetomidine in different settings, from its use in animal models of ischemia-reperfusion, and cardioprotective signaling pathways. In addition, pharmacokinetics and pharmacodynamics studies in obese subjects and the management of patients subjected to mechanical ventilation are described. Moreover, the clinical efficacy of delirium incidence in patients with indication of non-invasive ventilation is shown. Finally, the available evidence from DEX is described by a group of Chilean pharmacologists and clinicians who have worked for more than 10 years on DEX.
Collapse
Affiliation(s)
- Rodrigo L Castillo
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Unidad de Paciente Crítico, Hospital del Salvador, Santiago, Chile
| | - Mauricio Ibacache
- Programa de Farmacología y Toxicología & División de Anestesiología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio Cortínez
- Programa de Farmacología y Toxicología & División de Anestesiología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina Carrasco-Pozo
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Jorge G Farías
- Departmento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Francisco Salazar, Chile
| | - Rodrigo A Carrasco
- Departamento de Cardiología, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Patricio Vargas-Errázuriz
- Unidad de Paciente Crítico, Hospital del Salvador, Santiago, Chile.,Unidad de Paciente Crítico Adulto, Clínica Universidad de Los Andes, Santiago, Chile.,Unidad de Paciente Crítico, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Daniel Ramos
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Rafael Benavente
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Daniela Henríquez Torres
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Aníbal Méndez
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
15
|
Bach-Rojecky L, Vađunec D, Lozić M, Žunić K, Špoljar GG, Čutura T, Erceg D, Primorac D. Challenges in anesthesia personalization: resolving the pharmacogenomic puzzle. Per Med 2019; 16:511-525. [DOI: 10.2217/pme-2019-0056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Clinicians are witnessing differences in the doses required for induction and maintenance of anesthesia, as well as prolonged recovery in some patients. Predictable factors like patient characteristics, factors related to the procedure, pharmacological characteristics of anesthetics and adjunctive drugs, might explain some of the observed differences. However, the role of various polymorphisms of genes encoding for drugs’ molecular targets, transporters and metabolic enzymes can have a significant impact on anesthesia outcome, too. In the present paper, we critically discuss pharmacological characteristics of the most common drugs used in anesthesia, with a focus on the possible genetic background of unpredictable diversities in anesthesia outcomes.
Collapse
Affiliation(s)
- Lidija Bach-Rojecky
- Department of Pharmacology, University of Zagreb Faculty of Pharmacy & Biochemistry, Zagreb 10000, Croatia
| | - Dalia Vađunec
- Department of Pharmacology, University of Zagreb Faculty of Pharmacy & Biochemistry, Zagreb 10000, Croatia
| | - Marin Lozić
- Department of Anesthesia & ICU Care of Neurosurgical Patients, Clinic for Anesthesia & Intensive Care, University Clinical Hospital Zagreb, Zagreb 10000, Croatia
| | | | | | - Tomislav Čutura
- St. Catherine Specialty Hospital, Zagreb 10000 & Zabok 49210, Croatia
| | - Damir Erceg
- St. Catherine Specialty Hospital, Zagreb 10000 & Zabok 49210, Croatia
- Croatian Catholic University, Zagreb 10000, Croatia
- Srebrnjak Children's Hospital, Zagreb 10000, Croatia
- University Josip Juraj Strossmayer Faculty of Dental Medicine & Health, School of Medicine, Osijek 31000, Croatia
- University Josip Juraj Strossmayer School of Medicine, Osijek 31000, Croatia
| | - Dragan Primorac
- St. Catherine Specialty Hospital, Zagreb 10000 & Zabok 49210, Croatia
- University Josip Juraj Strossmayer Faculty of Dental Medicine & Health, School of Medicine, Osijek 31000, Croatia
- University Josip Juraj Strossmayer School of Medicine, Osijek 31000, Croatia
- Eberly College of Science, State College, Penn State University, PA 16802, USA
- The Henry C. Lee College of Criminal Justice & Forensic Sciences, University of New Haven, West Haven, CT 06516, USA
| |
Collapse
|
16
|
Pérez-Guillé MG, Toledo-López A, Rivera-Espinosa L, Alemon-Medina R, Murata C, Lares-Asseff I, Chávez-Pacheco JL, Gómez-Garduño J, Zamora Gutiérrez AL, Orozco-Galicia C, Ramírez-Morales K, Lugo-Goytia G. Population Pharmacokinetics and Pharmacodynamics of Dexmedetomidine in Children Undergoing Ambulatory Surgery. Anesth Analg 2019; 127:716-723. [PMID: 29782406 PMCID: PMC6110617 DOI: 10.1213/ane.0000000000003413] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Dexmedetomidine (DEX) is an α-2 adrenergic agonist with sedative and analgesic properties. Although not approved for pediatric use by the Food and Drug Administration, DEX is increasingly used in pediatric anesthesia and critical care. However, very limited information is available regarding the pharmacokinetics of DEX in children. The aim of this study was to investigate DEX pharmacokinetics and pharmacodynamics (PK-PD) in Mexican children 2-18 years of age who were undergoing outpatient surgical procedures. METHODS Thirty children 2-18 years of age with American Society of Anesthesiologists physical status score of I/II were enrolled in this study. DEX (0.7 μg/kg) was administered as a single-dose intravenous infusion. Venous blood samples were collected, and plasma DEX concentrations were analyzed with a combination of high-performance liquid chromatography and electrospray ionization-tandem mass spectrometry. Population PK-PD models were constructed using the Monolix program. RESULTS A 2-compartment model adequately described the concentration-time relationship. The parameters were standardized for a body weight of 70 kg by using an allometric model. Population parameters estimates were as follows: mean (between-subject variability): clearance (Cl) (L/h × 70 kg) = 20.8 (27%); central volume of distribution (V1) (L × 70 kg) = 21.9 (20%); peripheral volume of distribution (V2) (L × 70 kg) = 81.2 (21%); and intercompartmental clearance (Q) (L/h × 70 kg) = 75.8 (25%). The PK-PD model predicted a maximum mean arterial blood pressure reduction of 45% with an IC50 of 0.501 ng/ml, and a maximum heart rate reduction of 28.9% with an IC50 of 0.552 ng/ml. CONCLUSIONS Our results suggest that in Mexican children 2-18 years of age with American Society of Anesthesiologists score of I/II, the DEX dose should be adjusted in accordance with lower DEX clearance.
Collapse
Affiliation(s)
| | | | | | | | - Chiharu Murata
- Departamento de Metodología de la Investigación, Instituto Nacional de Pediatría, México
| | | | | | | | | | | | | | - Gustavo Lugo-Goytia
- Departamento de Anestesiología y Medicina Crítica, Instituto Nacional de Ciencias Médicas y Nutrición, México
| |
Collapse
|
17
|
Dutta A, Sethi N, Sood J, Panday BC, Gupta M, Choudhary P, Puri GD. The Effect of Dexmedetomidine on Propofol Requirements During Anesthesia Administered by Bispectral Index-Guided Closed-Loop Anesthesia Delivery System. Anesth Analg 2019; 129:84-91. [DOI: 10.1213/ane.0000000000003470] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
18
|
Zhu SJ, Wang KR, Zhang XX, Zhu SM. Relationship between genetic variation in the α 2A-adrenergic receptor and the cardiovascular effects of dexmedetomidine in the Chinese Han population. J Zhejiang Univ Sci B 2019; 20:598-604. [PMID: 31168973 DOI: 10.1631/jzus.b1800647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
There are differences in individual cardiovascular responses to the administration of dexmedetomidine, a highly selective α2A-adrenergic receptor (ADRA2A) agonist. The aim of this study was to investigate ADRA2A gene polymorphisms in the Chinese Han population and their association with the cardiovascular response to intravenous dexmedetomidine infusion. Sixty elective surgery patients of Chinese Han nationality were administered 1 µg/kg dexmedetomidine intravenously over 10 min as a premedication. ADRA2A C-1291G and A1780G polymorphism status was determined in these patients, and their relationships to changes in blood pressure and heart rate after dexmedetomidine administration were analyzed. There were neither significant differences in systolic or diastolic blood pressure changes in individuals with different A1780G and C-1291G genotypes after dexmedetomidine administration, nor in heart rates among the different A1780G genotypes. However, there were significant differences in changes in heart rates in patients with different C-1291G genotypes. There were no significant differences in the sedative effects of dexmedetomidine among different A1780G and C-1291G genotypes. Logistic regression revealed that the C-1291G polymorphism was associated with differential decreases in heart rate after intravenous infusion of dexmedetomidine. These findings indicate that the ADRA2A C-1291G polymorphism can affect heart rate changes in patients after intravenous infusion of dexmedetomidine.
Collapse
Affiliation(s)
- Shao-Jun Zhu
- Department of Anesthesiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Kui-Rong Wang
- Department of Anesthesiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xiong-Xin Zhang
- Department of Anesthesiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Sheng-Mei Zhu
- Department of Anesthesiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
19
|
Zhou S, Skaar DJ, Jacobson PA, Huang RS. Pharmacogenomics of Medications Commonly Used in the Intensive Care Unit. Front Pharmacol 2018; 9:1436. [PMID: 30564130 PMCID: PMC6289166 DOI: 10.3389/fphar.2018.01436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/20/2018] [Indexed: 12/11/2022] Open
Abstract
In the intensive care unit (ICU) setting, where highly variable and insufficient drug efficacies, as well as frequent and unpredictable adverse drug reactions (ADRs) occur, pharmacogenomics (PGx) offers an opportunity to improve health outcomes. However, PGx has not been fully evaluated in the ICU, partly due to lack of knowledge of how genetic markers may affect drug therapy. To fill in this gap, we conducted a review to summarize the PGx information for the medications commonly encountered in the ICU.
Collapse
Affiliation(s)
- Shuqin Zhou
- Department of Emergency and Critical Care Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.,Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Debra J Skaar
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Pamala A Jacobson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - R Stephanie Huang
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
20
|
Wang L, Wang S, Qi J, Yu R, Zhuang J, Zhuang B, Lou Y, Ruan J, Ye H, Lin F. Impact of CYP2A6 gene polymorphism on the pharmacokinetics of dexmedetomidine for premedication. Expert Rev Clin Pharmacol 2018; 11:917-922. [PMID: 30092666 DOI: 10.1080/17512433.2018.1510312] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
BACKGROUND Dexmedetomidine is a widely used sedative in clinic, which is mainly metabolized by cytochrome P450 2A6 (CYP2A6). Dexmedetomidine was rarely reported for off-label usage of premedication, but lacking relevant pharmacokinetic investigations. Therefore, our study determined the dexmedetomidine pharmacokinetics of CYP2A6*4 allele in Chinese patients pretreated with dexmedetomidine whose mutation frequency of CYP2A6*4 are high, in order to provide clinical references. METHODS Thirty-one elective surgery patients received premedication with 0.5 μg/kg dexmedetomidine via intravenous pump. Their plasma concentrations at multiple time-points and polymorphism of CYP2A6*4 were determined and statistically analyzed. RESULTS 9 patients were *1/*4 or *4/*4, and 22 patients were *1/*1. The main pharmacokinetic parameters were area under curve (AUC) 1396.19 ± 332.47h· ng· l-1, peak blood concentration (Cmax) 495.50 ± 104.90ng· l-1, distribution volume (V) 0.68 ± 0.20 L/kg, clearance (CL) 0.38 ± 0.11 L/h/kg, distribution half-life (t1/2α) 0.05 ± 0.01h, elimination half-life (t1/2β) 2.53 ± 0.04h. No significant pharmacokinetic differences were found among CYP2A6*1/*1, *1/*4, and *4/*4 patients. CONCLUSIONS In Chinese patients pretreated with dexmedetomidine, T1/2β was consistent with that published, but T1/2α, V and Cl were lower. It was unnecessary to consider the mutation when developing the precision regimen of dexmedetomidine.
Collapse
Affiliation(s)
- Ling Wang
- a Department of Pharmacy , Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University , Fuzhou , China
| | - Shaoming Wang
- a Department of Pharmacy , Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University , Fuzhou , China
| | - Juan Qi
- b Second Department of Anesthesiology , Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University , Fuzhou , China
| | - Rongguo Yu
- c Surgery Intensive Care Unit , Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University , Fuzhou , China
| | - Jie Zhuang
- a Department of Pharmacy , Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University , Fuzhou , China
| | - Boyang Zhuang
- d Center for Certification and Evaluation , Fujian Food and Drug Administration , Fuzhou , China
| | - Yongming Lou
- e Chemical Laboratory , Fujian Institute for Food and Drug Quality Control , Fuzhou , China
| | - Junshan Ruan
- a Department of Pharmacy , Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University , Fuzhou , China
| | - Hong Ye
- a Department of Pharmacy , Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University , Fuzhou , China
| | - Fangfang Lin
- a Department of Pharmacy , Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University , Fuzhou , China
| |
Collapse
|
21
|
Zheng Z, Zhang S, Ma W, Zhang L, Huang L, Huang W, Huang M, Wang Z, Li J. Determination of dexmedetomidine by UHPLC–MS/MS and its application to evaluate the effect of dexmedetomidine concentration on the target-controlled infusion concentration of propofol. J Pharm Biomed Anal 2018; 154:438-443. [DOI: 10.1016/j.jpba.2018.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/15/2018] [Accepted: 03/07/2018] [Indexed: 12/26/2022]
|
22
|
Abstract
Dexmedetomidine is an α2-adrenoceptor agonist with sedative, anxiolytic, sympatholytic, and analgesic-sparing effects, and minimal depression of respiratory function. It is potent and highly selective for α2-receptors with an α2:α1 ratio of 1620:1. Hemodynamic effects, which include transient hypertension, bradycardia, and hypotension, result from the drug’s peripheral vasoconstrictive and sympatholytic properties. Dexmedetomidine exerts its hypnotic action through activation of central pre- and postsynaptic α2-receptors in the locus coeruleus, thereby inducting a state of unconsciousness similar to natural sleep, with the unique aspect that patients remain easily rousable and cooperative. Dexmedetomidine is rapidly distributed and is mainly hepatically metabolized into inactive metabolites by glucuronidation and hydroxylation. A high inter-individual variability in dexmedetomidine pharmacokinetics has been described, especially in the intensive care unit population. In recent years, multiple pharmacokinetic non-compartmental analyses as well as population pharmacokinetic studies have been performed. Body size, hepatic impairment, and presumably plasma albumin and cardiac output have a significant impact on dexmedetomidine pharmacokinetics. Results regarding other covariates remain inconclusive and warrant further research. Although initially approved for intravenous use for up to 24 h in the adult intensive care unit population only, applications of dexmedetomidine in clinical practice have been widened over the past few years. Procedural sedation with dexmedetomidine was additionally approved by the US Food and Drug Administration in 2003 and dexmedetomidine has appeared useful in multiple off-label applications such as pediatric sedation, intranasal or buccal administration, and use as an adjuvant to local analgesia techniques.
Collapse
|
23
|
Rolle A, Paredes S, Cortínez LI, Anderson BJ, Quezada N, Solari S, Allende F, Torres J, Cabrera D, Contreras V, Carmona J, Ramírez C, Oliveros AM, Ibacache M. Dexmedetomidine metabolic clearance is not affected by fat mass in obese patients. Br J Anaesth 2018; 120:969-977. [PMID: 29661414 DOI: 10.1016/j.bja.2018.01.040] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/28/2017] [Accepted: 01/27/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Obesity has been associated with reduced dexmedetomidine clearance, suggesting impaired hepatic function or reduced hepatic blood flow. The aim of this study was to clarify the effect of obesity in dexmedetomidine metabolic clearance. METHODS Forty patients, ASA I-III, 18-60 yr old, weighing 47-126 kg, scheduled for abdominal laparoscopic surgery, were enrolled. Anaesthetic agents (propofol, remifentanil, and dexmedetomidine) were dosed based on lean body weight measured by dual X-ray absorptiometry. Serial venous samples were drawn during and after dexmedetomidine infusion. A pharmacokinetic analysis was undertaken using non-linear mixed-effect models. In the modelling approach, the total body weight, lean body weight, and adjusted body weight were first tested as size descriptors for volumes and clearances. Hepatic blood flow, liver histopathology, liver enzymes, and gene expression of metabolic enzymes (UGT2B10 and UGT1A4) were tested as covariates of dexmedetomidine metabolic clearance. A decrease in NONMEM objective function value (ΔOFV) of 3.84 points, for an added parameter, was considered significant at the 0.05 level. RESULTS A total of 637 dexmedetomidine serum samples were obtained. A two-compartmental model scaled to measured lean weight adequately described the dexmedetomidine pharmacokinetics. Liver blood flow was a covariate for dexmedetomidine clearance (ΔOFV=-5.878). Other factors, including fat mass, histopathological damage, and differential expression of enzymes, did not affect the dexmedetomidine clearance in the population studied (ΔOFV<3.84). CONCLUSIONS We did not find a negative influence of obesity in dexmedetomidine clearance when doses were adjusted to lean body weight. Liver blood flow showed a significant effect on dexmedetomidine clearance. CLINICAL TRIAL REGISTRATION NCT02557867.
Collapse
Affiliation(s)
- A Rolle
- División de Anestesiología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - S Paredes
- División de Anestesiología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - L I Cortínez
- División de Anestesiología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - B J Anderson
- Department of Anaesthesiology, University of Auckland, Auckland, New Zealand
| | - N Quezada
- Departamento de Cirugía Digestiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - S Solari
- Departamento de Laboratorio Clínico, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - F Allende
- Departamento de Laboratorio Clínico, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - J Torres
- Departamento de Anatomía Patológica, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - D Cabrera
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Ciencias Químicas y Biológicas, Facultad de Salud, Universidad Bernardo O'Higgins, Santiago, Chile
| | - V Contreras
- División de Anestesiología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - J Carmona
- División de Anestesiología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - C Ramírez
- División de Anestesiología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A M Oliveros
- División de Anestesiología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - M Ibacache
- División de Anestesiología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
24
|
Xie S, Ma W, Guo Q, Liu J, Li W, McLeod HL, He Y. The pharmacogenetics of medications used in general anesthesia. Pharmacogenomics 2018; 19:285-298. [PMID: 29318929 DOI: 10.2217/pgs-2017-0168] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
General anesthesia is a state of unconsciousness, amnesia, analgesia and akinesia induced by drugs including opioids, hypnotic-sedative agents, muscle relaxants and antiemetics. Clinical and genetic factors are reported to influence the efficacy and side effects of these agents. Based on the evidence, clinical action is needed to improve clinical outcomes. This review summarizes the latest knowledge with regards to the pharmacogenetics of anesthetics and general anesthesia related complications.
Collapse
Affiliation(s)
- Shangchen Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, P.R. China
| | - Wenjuan Ma
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Jie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, P.R. China
| | - Wei Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, P.R. China
| | - Howard L McLeod
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, P.R. China
- Moffitt Cancer Center, DeBartolo Family Personalized Medicine Institute, Tampa, FL 33601, USA
| | - Yijing He
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, P.R. China
- Moffitt Cancer Center, DeBartolo Family Personalized Medicine Institute, Tampa, FL 33601, USA
| |
Collapse
|
25
|
Pharmacokinetic and pharmacodynamics of intravenous dexmedetomidine in morbidly obese patients undergoing laparoscopic surgery. J Anesth 2017; 31:813-820. [DOI: 10.1007/s00540-017-2399-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/04/2017] [Indexed: 01/02/2023]
|
26
|
Liu HC, Lian QQ, Wu FF, Wang CY, Sun W, Zheng LD, Schüttler J, Ihmsen H. Population Pharmacokinetics of Dexmedetomidine After Short Intravenous Infusion in Chinese Children. Eur J Drug Metab Pharmacokinet 2017; 42:201-211. [PMID: 27037817 DOI: 10.1007/s13318-016-0333-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Dexmedetomidine is a highly selective alpha2-adrenoceptor agonist with sedative and analgesic properties which is also used in pediatric anesthesia. Although the pharmacokinetics of dexmedetomidine have been studied in pediatric patients, there are no data for Chinese children available. As alterations in pharmacokinetics due to ethnicity cannot be ruled out, it was the aim of this study to characterize the pharmacokinetics of dexmedetomidine in Chinese pediatric patients. METHODS Thirty-nine children aged 1-9 years undergoing surgery were enrolled in the study. Dexmedetomidine was administered as short intravenous infusion of 1-2 µg/kg in 10 min. Venous blood samples were drawn until 480 min after stopping of infusion. Dexmedetomidine plasma concentrations were measured with high-performance liquid chromatography and mass spectrometry. Pharmacokinetic modeling was performed by population analysis using linear compartment models. RESULTS Data of 36 patients (age 1-9 years, weight 10-27 kg) were analyzed. The pharmacokinetics of dexmedetomidine were best described by a two-compartment model with an allometric power model and estimates standardized to 70 kg body weight. The population estimates (95 % CI) per 70 kg bodyweight were: clearance 36.2 (33.3-41.1) l/h, central volume of distribution 84.3 (70.3-91.4) l, intercompartmental clearance 82.8 (63.6-136.6) l/h, peripheral volume of distribution 114 (95-149) l, and terminal half-life 4.4 (3.6-5.3) h. Age did not show any influence on weight-adjusted parameters. CONCLUSIONS Chinese children showed a similar clearance, but larger volumes of distribution and longer terminal half-life when compared to studies in Caucasians. TRIAL REGISTRATION ChiCTR-OPC-14005659.
Collapse
Affiliation(s)
- Hua-Cheng Liu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing-Quan Lian
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fei-Fei Wu
- Department of Anesthesiology, Anqing First People's Hospital of Anhui Province, Anqing, China
| | - Cheng-Yu Wang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Sun
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li-Dan Zheng
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jürgen Schüttler
- Department of Anesthesiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstrasse 12, 91054, Erlangen, Germany
| | - Harald Ihmsen
- Department of Anesthesiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstrasse 12, 91054, Erlangen, Germany.
| |
Collapse
|
27
|
Li A, Yuen VMY, Goulay-Dufay S, Kwok PCL. Pharmacokinetics and pharmacodynamics of dexmedetomidine. Drug Dev Ind Pharm 2016; 42:1917-1927. [PMID: 27595299 DOI: 10.1080/03639045.2016.1232727] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Dexmedetomidine is an alpha-2 adrenoceptor agonist and has been used as a general anesthetic, sedative and analgesic for about 30 years. The aim of this paper is to review the pharmacokinetics and pharmacodynamics of dexmedetomidine, evaluate physiological factors that may affect the pharmacokinetics of dexmedetomidine, and summarize the pharmacodynamics of dexmedetomidine at different plasma levels. The pharmacokinetic parameters reported in previous studies according to noncompartmental analyses or population modeling results are compared. We concluded that the pharmacokinetic profile can be adequately described by a two-compartment model in population pharmacokinetic modeling. Body weight, height, albumin level, cardiac output, disease condition and other factors were considered to have significant influence on the clearance and/or distribution volume in different population pharmacokinetic models. The pharmacological effects of dexmedetomidine, such as sedation, heart rate reduction and biphasic change of blood pressure, vary at different plasma levels. These findings provide a reference for individualizing the dose of dexmedetomidine and achieving the desired pharmacological effects in clinical applications.
Collapse
Affiliation(s)
- Aiwei Li
- a Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine , The University of Hong Kong , Pokfulam , Hong Kong SAR , China
| | - Vivian Man Ying Yuen
- b Department of Anesthesiology , University of Hong Kong Shenzhen Hospital , Futian , Guangdong , Shenzhen , China
| | - Sophie Goulay-Dufay
- c Département Laboratoires, AGEPS , Assistance Publique des Hôpitaux de Paris , Paris , France
| | - Philip Chi Lip Kwok
- a Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine , The University of Hong Kong , Pokfulam , Hong Kong SAR , China
| |
Collapse
|
28
|
Abstract
OBJECTIVE To characterize fentanyl population pharmacokinetics in patients with critical illness and identify patient characteristics associated with altered fentanyl concentrations. DESIGN Prospective cohort study. SETTING Medical and surgical ICUs in a large tertiary care hospital in the United States. PATIENTS Patients with acute respiratory failure and/or shock who received fentanyl during the first 5 days of their ICU stay. MEASUREMENTS AND MAIN RESULTS We collected clinical and hourly drug administration data and measured fentanyl concentrations in plasma collected once daily for up to 5 days after enrollment. Among 337 patients, the mean duration of infusion was 58 hours at a median rate of 100 μg/hr. Using a nonlinear mixed-effects model implemented by NONMEM, we found that fentanyl pharmacokinetics were best described by a two-compartment model in which weight, severe liver disease, and congestive heart failure most affected fentanyl concentrations. For a patient population with a mean weight of 92 kg and no history of severe liver disease or congestive heart failure, the final model, which performed well in repeated 10-fold cross-validation, estimated total clearance, intercompartmental clearance (Q), and volumes of distribution for the central (V1) and peripheral compartments (V2) to be 35 L/hr (95% CI, 32-39 L/hr), 55 L/hr (95% CI, 42-68 L/hr), 203 L (95% CI, 140-266 L), and 523 L (95% CI, 428-618 L), respectively. Severity of illness was marginally associated with fentanyl pharmacokinetics but did not improve the model fit after liver and heart diseases were included. CONCLUSIONS In this study, fentanyl pharmacokinetics during critical illness were strongly influenced by severe liver disease, congestive heart failure, and weight, factors that should be considered when dosing fentanyl in the ICU. Future studies are needed to determine if data-driven fentanyl dosing algorithms can improve outcomes for ICU patients.
Collapse
|
29
|
Li C, Li Y, Wang K, Kong X. Comparative Evaluation of Remifentanil and Dexmedetomidine in General Anesthesia for Cesarean Delivery. Med Sci Monit 2015; 21:3806-13. [PMID: 26638888 PMCID: PMC4676355 DOI: 10.12659/msm.895209] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Use of remifentanil and dexmedetomidine in general anesthesia for cesarean section have been described. This study was designed to evaluate the effects of remifentanil and dexmedetomidine on maternal hemodynamics and bispectral index, and neonatal outcomes in elective caesarean delivery. Material/Methods Forty-four women undergoing elective cesarean delivery with ASA I or II and term or near-term singleton pregnancies were randomly assigned to receive remifentanil at a loading dose of 2 μg/kg over 10 min followed by a continuous infusion of 2 μg/kg/h until about 6 min before fetal delivery (Group REM), or dexmedetomidine at a loading dose of 0.4 μg/kg over 10 min followed by a continuous infusion of 0.4 μg/kg/h until about 6 min before fetal delivery (Group DEX). Maternal hemodynamics and BIS values were recorded. Neonatal effects were assessed using Apgar scores and umbilical cord blood gas analysis. Results Mean arterial pressure (MAP) increased after intubation in both groups, and the change magnitude of the MAP was higher in Group DEX (P<0.05). Patients in Group DEX had a lower BIS value at recovery and consumed less propofol during surgery (P<0.05). The incidences of neonatal resuscitation at 1 min were 81.8% in Group REM and 54.5% in Group DEX (P=0.052). There was no significant difference in either group in Apgar scores at 1 and 5 min and umbilical cord blood gas values. Conclusions Both remifentanil and dexmedetomidine are effective to blunt hemodynamic responses to intubation and also seem safe for neonates at the administrated doses, but remifentanil still has the potential to cause neonatal transient respiratory depression.
Collapse
Affiliation(s)
- Chengwen Li
- Department of Anesthesiology, Jining No. 1 People's Hospital, Jining, Shandong, China (mainland)
| | - Yandong Li
- Department of Anesthesiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| | - Kun Wang
- Department of Anesthesiology, Jining No. 1 People's Hospital, Jining, Shandong, China (mainland)
| | - Xiangang Kong
- Department of Anesthesiology, Jining No. 1 People's Hospital, Jining, Shandong, China (mainland)
| |
Collapse
|
30
|
Dexmedetomidine pharmacokinetics in the obese. Eur J Clin Pharmacol 2015; 71:1501-8. [DOI: 10.1007/s00228-015-1948-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 09/14/2015] [Indexed: 10/23/2022]
|
31
|
Abstract
Dexmedetomidine (Dexdor(®)) is a highly selective α2-adrenoceptor agonist. It has sedative, analgesic and opioid-sparing effects and is suitable for short- and longer-term sedation in an intensive care setting. In the randomized, double-blind, multicentre MIDEX and PRODEX trials, longer-term sedation with dexmedetomidine was noninferior to midazolam and propofol in terms of time spent at the target sedation range, as well as being associated with a shorter time to extubation than midazolam or propofol, and a shorter duration of mechanical ventilation than midazolam. Patients receiving dexmedetomidine were also easier to rouse, more co-operative and better able to communicate than patients receiving midazolam or propofol. Dexmedetomidine had beneficial effects on delirium in some randomized, controlled trials (e.g. patients receiving dexmedetomidine were less likely to experience delirium than patients receiving midazolam, propofol or remifentanil and had more delirium- and coma-free days than patients receiving lorazepam). Intravenous dexmedetomidine had an acceptable tolerability profile; hypotension, hypertension and bradycardia were the most commonly reported adverse reactions. In conclusion, dexmedetomidine is an important option for sedation in the intensive care setting.
Collapse
Affiliation(s)
- Gillian M Keating
- Springer, Private Bag 65901, Mairangi Bay 0754, Auckland, New Zealand,
| |
Collapse
|
32
|
Ranheim B, Risberg ÅI, Spadavecchia C, Landsem R, Haga HA. The pharmacokinetics of dexmedetomidine administered as a constant rate infusion in horses. J Vet Pharmacol Ther 2014; 38:93-6. [PMID: 25229603 DOI: 10.1111/jvp.12157] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 07/17/2014] [Indexed: 12/26/2022]
Abstract
Dexmedetomidine, the most selective α2-adrenoceptor agonist in clinical use, is increasingly being used in both conscious and anaesthetized horses; however, the pharmacokinetics and sedative effects of this drug administered alone as an infusion are not previously described in horses. Seven horses received an infusion of 8 μg dexmedetomidine/kg/h for 150 min, venous blood samples were collected, and dexmedetomidine concentrations were assayed using liquid chromatography-mass spectrometry (LC/MS) and analyzed using noncompartmental pharmacokinetic analysis. Sedation was scored as the distance from the lower lip of the horse to the ground measured in centimetre. The harmonic mean (SD) plasma elimination half-life (Lambda z half-life) for dexmedetomidine was 20.9 (5.1) min, clearance (Cl) was 0.3 (0.20) L/min/kg, and volume of distribution at steady-state (Vdss ) was 13.7 (7.9) L/kg. There was a considerable individual variation in the concentration of dexmedetomidine vs. time profile. The level of sedation covaried with the plasma concentration of dexmedetomidine. This implies that for clinical use of dexmedetomidine constant rate infusion in conscious horses, infusion rates can be easily adjusted to effect, and this is preferable to an infusion at a predetermined value.
Collapse
Affiliation(s)
- B Ranheim
- Faculty of Veterinary Medicine and Biosciences, The Norwegian University of Life Sciences, Oslo, Norway
| | | | | | | | | |
Collapse
|
33
|
Smithburger PL, Smith RB, Kane-Gill SL, Empey PE. Patient predictors of dexmedetomidine effectiveness for sedation in intensive care units. Am J Crit Care 2014; 23:160-5. [PMID: 24585165 DOI: 10.4037/ajcc2014678] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
BACKGROUND Dexmedetomidine, a selective α2-adrenergic receptor agonist, is increasingly used as a sedative in intensive care despite variations in patients' responses. OBJECTIVES To determine the effectiveness of dexmedetomidine as a sedative and specific patient characteristics that play a role in adequate sedation with dexmedetomidine. METHODS A 6-month, pilot, prospective observational study was performed in a medical intensive care unit at an academic medical center. Patients receiving dexmedetomidine were followed up until use of the drug was stopped and they were classified as nonresponders or responders. Effective sedation was defined as a score of 3 to 4 on the Sedation Agitation Scale after the administration of dexmedetomidine. Patient characteristics, laboratory values, home and inpatient medications, and dexmedetomidine dosing information were collected to identify predictors of clinical response. RESULTS During the 6-month study period, 38 patients received dexmedetomidine. The drug was ineffective as a sedative in 19 patients (50%) and effective in 11 (29%). Effectiveness could not be assessed in 8 patients because of clinical confounders. According to standard multiple logistic regression analysis, successful sedation was more likely in patients with a lower score on the Acute Physiology and Chronic Health Evaluation II (Odds Ratio [OR] 0.81; 95% CI, -0.39 to -0.03) and patients who took antidepressants at home (OR 10.27; 95% CI, 0.23 to 4.43) than in patients who had a higher score or did not take antidepressants at home. CONCLUSIONS Effective sedation with dexmedetomidine is variable.
Collapse
Affiliation(s)
- Pamela L. Smithburger
- Pamela L. Smithburger and Philip E. Empey are assistant professors, Randall B. Smith is senior associate dean, and Sandra L. Kane-Gill is an associate professor at University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
| | - Randall B. Smith
- Pamela L. Smithburger and Philip E. Empey are assistant professors, Randall B. Smith is senior associate dean, and Sandra L. Kane-Gill is an associate professor at University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
| | - Sandra L. Kane-Gill
- Pamela L. Smithburger and Philip E. Empey are assistant professors, Randall B. Smith is senior associate dean, and Sandra L. Kane-Gill is an associate professor at University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
| | - Philip E. Empey
- Pamela L. Smithburger and Philip E. Empey are assistant professors, Randall B. Smith is senior associate dean, and Sandra L. Kane-Gill is an associate professor at University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
| |
Collapse
|
34
|
Välitalo PA, Ahtola-Sätilä T, Wighton A, Sarapohja T, Pohjanjousi P, Garratt C. Population pharmacokinetics of dexmedetomidine in critically ill patients. Clin Drug Investig 2014; 33:579-87. [PMID: 23839483 PMCID: PMC3717151 DOI: 10.1007/s40261-013-0101-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background and Objectives Although the pharmacokinetics of dexmedetomidine in healthy volunteers have been studied, there are limited data about the pharmacokinetics of long-term administration of dexmedetomidine in critically ill patients. Methods This population pharmacokinetic analysis was performed to quantify the pharmacokinetics of dexmedetomidine in critically ill patients following infusions up to 14 days in duration. The data consisted of three phase III studies (527 patients with sparse blood sampling, for a total of 2,144 samples). Covariates were included in a full random-effects covariate model and the most important covariate relationships were tested separately. The linearity of dexmedetomidine clearance was evaluated by observing steady-state plasma concentrations acquired at various infusion rates. Results The data were adequately described with a one-compartment model. The clearance of dexmedetomidine was 39 (95 % CI 37–41) L/h and volume of distribution 104 (95 % CI 93–115) L. Both clearance and volume of distribution were highly variable between patients (coefficients of variation of 62 and 57 %, respectively), which highlights the importance of dose titration by response. Covariate analysis showed a strong correlation between body weight and clearance of dexmedetomidine. The clearance of dexmedetomidine was constant in the dose range 0.2–1.4 μg/kg/h. Conclusions The pharmacokinetics of dexmedetomidine are dose-proportional in prolonged infusions when dosing rates of 0.2–1.4 μg/kg/h, recommended by the Dexdor® summary of product characteristics, are used.
Collapse
Affiliation(s)
- Pyry Antti Välitalo
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland, PO Box 1624, 70211, Kuopio, Finland.
| | | | | | | | | | | |
Collapse
|
35
|
Holliday SF, Kane-Gill SL, Empey PE, Buckley MS, Smithburger PL. Interpatient variability in dexmedetomidine response: a survey of the literature. ScientificWorldJournal 2014; 2014:805013. [PMID: 24558330 PMCID: PMC3914598 DOI: 10.1155/2014/805013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 10/22/2013] [Indexed: 12/28/2022] Open
Abstract
Fifty-five thousand patients are cared for in the intensive care unit (ICU) daily with sedation utilized to reduce anxiety and agitation while optimizing comfort. The Society of Critical Care Medicine (SCCM) released updated guidelines for management of pain, agitation, and delirium in the ICU and recommended nonbenzodiazepines, such as dexmedetomidine and propofol, as first line sedation agents. Dexmedetomidine, an alpha-2 agonist, offers many benefits yet its use is mired by the inability to consistently achieve sedation goals. Three hypotheses including patient traits/characteristics, pharmacokinetics in critically ill patients, and clinically relevant genetic polymorphisms that could affect dexmedetomidine response are presented. Studies in patient traits have yielded conflicting results regarding the role of race yet suggest that dexmedetomidine may produce more consistent results in less critically ill patients and with home antidepressant use. Pharmacokinetics of critically ill patients are reported as similar to healthy individuals yet wide, unexplained interpatient variability in dexmedetomidine serum levels exist. Genetic polymorphisms in both metabolism and receptor response have been evaluated in few studies, and the results remain inconclusive. To fully understand the role of dexmedetomidine, it is vital to further evaluate what prompts such marked interpatient variability in critically ill patients.
Collapse
Affiliation(s)
- Samantha F. Holliday
- University of Pittsburgh School of Pharmacy, 3501 Terrace Street, Pittsburgh, PA 15261, USA
| | - Sandra L. Kane-Gill
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, 3501 Terrace Street, Pittsburgh, PA 15261, USA
| | - Philip E. Empey
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, 3501 Terrace Street, Pittsburgh, PA 15261, USA
| | - Mitchell S. Buckley
- Banner Good Samaritan Medical Center, Department of Pharmacy, 1111 E. McDowell Road, Phoenix, AZ 85006, USA
| | - Pamela L. Smithburger
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, 3501 Terrace Street, Pittsburgh, PA 15261, USA
| |
Collapse
|
36
|
MacLaren R, Krisl JC, Cochrane RE, Mueller SW. A case-based approach to the practical application of dexmedetomidine in critically ill adults. Pharmacotherapy 2013; 33:165-86. [PMID: 23386596 DOI: 10.1002/phar.1175] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dexmedetomidine is a selective α(2) -adrenoceptor agonist that offers unique sedation because patients are readily awakened while administration continues and the drug does not suppress the respiratory center. Limitations of use include higher acquisition cost, inability to produce deep sedation, and bradycardia and hypotension. Using a case-based approach, the purpose of this review was to qualitatively assess the role of dexmedetomidine in the care of the critically ill and in the management of alcohol withdrawal, and to formulate recommendations regarding its clinical application. Sixty-six studies were identified that investigated dexmedetomidine for the provision of sedation. These studies were heterogeneous in design and patient populations; most investigated patients did not require heavy sedation, and few used propofol as the comparator. In general, though, the aggregate results of all studies demonstrate that dexmedetomidine provides comfort, possibly shortens the duration of mechanical ventilation to facilitate extubation, reduces the occurrence of acute brain dysfunction, and facilitates communication, but the drug is associated with hemodynamic instability and requires the supplemental use of traditional sedative and analgesic agents. These outcomes need to be substantiated in additional studies that include assessments of cost-effectiveness. Dexmedetomidine should be considered when patients require mild to moderate levels of sedation of short to intermediate time frames, and they qualify for daily awakenings with traditional sedative therapies. The data for dexmedetomidine in relation to alcohol withdrawal are limited to 12 retrospective reports representing a total of 127 patients. Its role for this indication requires further study, but it may be considered as adjunctive therapy when clinicians are concerned about respiratory suppression associated with escalating doses of γ-aminobutyric acid agonists. Regardless of the indication for dexmedetomidine, the practitioner must closely monitor patient comfort and the occurrence of hemodynamic deviations with the realization that as-needed administration of traditional sedatives and analgesics will be required and some degree of bradycardia and hypotension expected but intervention rarely required.
Collapse
Affiliation(s)
- Robert MacLaren
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado 80045, USA.
| | | | | | | |
Collapse
|