1
|
Saravanapandian V, Madani M, Nichols I, Vincent S, Dover M, Dikeman D, Philpot BD, Takumi T, Colwell CS, Jeste S, Paul KN, Golshani P. Sleep EEG signatures in mouse models of 15q11.2-13.1 duplication (Dup15q) syndrome. J Neurodev Disord 2024; 16:39. [PMID: 39014349 PMCID: PMC11251350 DOI: 10.1186/s11689-024-09556-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/02/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Sleep disturbances are a prevalent and complex comorbidity in neurodevelopmental disorders (NDDs). Dup15q syndrome (duplications of 15q11.2-13.1) is a genetic disorder highly penetrant for NDDs such as autism and intellectual disability and it is frequently accompanied by significant disruptions in sleep patterns. The 15q critical region harbors genes crucial for brain development, notably UBE3A and a cluster of gamma-aminobutyric acid type A receptor (GABAAR) genes. We previously described an electrophysiological biomarker of the syndrome, marked by heightened beta oscillations (12-30 Hz) in individuals with Dup15q syndrome, akin to electroencephalogram (EEG) alterations induced by allosteric modulation of GABAARs. Those with Dup15q syndrome exhibited increased beta oscillations during the awake resting state and during sleep, and they showed profoundly abnormal NREM sleep. This study aims to assess the translational validity of these EEG signatures and to delve into their neurobiological underpinnings by quantifying sleep physiology in chromosome-engineered mice with maternal (matDp/ + mice) or paternal (patDp/ + mice) inheritance of the full 15q11.2-13.1-equivalent duplication, and mice with duplication of just the UBE3A gene (Ube3a overexpression mice; Ube3a OE mice) and comparing the sleep metrics with their respective wildtype (WT) littermate controls. METHODS We collected 48-h EEG/EMG recordings from 35 (23 male, 12 female) 12-24-week-old matDp/ + , patDp/ + , Ube3a OE mice, and their WT littermate controls. We quantified baseline sleep, sleep fragmentation, spectral power dynamics during sleep states, and recovery following sleep deprivation. Within each group, distinctions between Dup15q mutant mice and WT littermate controls were evaluated using analysis of variance (ANOVA) and student's t-test. The impact of genotype and time was discerned through repeated measures ANOVA, and significance was established at p < 0.05. RESULTS Our study revealed that across brain states, matDp/ + mice mirrored the elevated beta oscillation phenotype observed in clinical EEGs from individuals with Dup15q syndrome. Time to sleep onset after light onset was significantly reduced in matDp/ + and Ube3a OE mice. However, NREM sleep between Dup15q mutant and WT littermate mice remained unaltered, suggesting a divergence from the clinical presentation in humans. Additionally, while increased beta oscillations persisted in matDp/ + mice after 6-h of sleep deprivation, recovery NREM sleep remained unaltered in all groups, thus suggesting that these mice exhibit resilience in the fundamental processes governing sleep-wake regulation. CONCLUSIONS Quantification of mechanistic and translatable EEG biomarkers is essential for advancing our understanding of NDDs and their underlying pathophysiology. Our study of sleep physiology in the Dup15q mice underscores that the beta EEG biomarker has strong translational validity, thus opening the door for pre-clinical studies of putative drug targets, using the biomarker as a translational measure of drug-target engagement. The unaltered NREM sleep may be due to inherent differences in neurobiology between mice and humans. These nuanced distinctions highlight the complexity of sleep disruptions in Dup15q syndrome and emphasize the need for a comprehensive understanding that encompasses both shared and distinct features between murine models and clinical populations.
Collapse
Affiliation(s)
- Vidya Saravanapandian
- Department of Neurology and Semel Institute for Neuroscience, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA.
| | - Melika Madani
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - India Nichols
- Department of Biology, Spelman College, 350 Spelman Lane, Atlanta, GA, 30314, USA
| | - Scott Vincent
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Mary Dover
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Dante Dikeman
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Benjamin D Philpot
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities, UNC-Chapel Hill, NC, 27599, USA
| | - Toru Takumi
- Kobe University School of Medicine, Chuo, Kobe, 650-0017, Japan
| | - Christopher S Colwell
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Psychiatry & Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Shafali Jeste
- Children's Hospital Los Angeles, 4650 Sunset Blvd, MS 82, Los Angeles, CA, 90027, USA
| | - Ketema N Paul
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Peyman Golshani
- Department of Neurology and Semel Institute for Neuroscience, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
- West Los Angeles VA Medical Center, 11301 Wilshire Blvd, Los Angeles, CA, 90073, USA
| |
Collapse
|
2
|
Wang L, Wang H, Tang W, Tang L, Xu Y, Xiong L. Comparison of intranasal dexmedetomidine alone and dexmedetomidine-chloral hydrate combination sedation for electroencephalography in children: A large retrospective cohort study and propensity score-matched analysis. Heliyon 2024; 10:e32236. [PMID: 38873690 PMCID: PMC11170184 DOI: 10.1016/j.heliyon.2024.e32236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024] Open
Abstract
Aim To compare the safety and efficacy of intranasal high-dose dexmedetomidine (DEX) versus a combination of intranasal low-dose dexmedetomidine and oral chloral hydrate (DEX-CH) sedation during electroencephalography (EEG) in children. Methods Unadjusted analysis, 1:1 propensity score matching (PSM), and inverse probability of treatment weighting (IPTW) were used to compare the sedation success rate, adverse effects, onset time, and recovery time of these two sedation methods for 6967 children who underwent EEG. Results A total of 6967 children were enrolled in this study, of whom 846 (12.1 %) underwent DEX intranasal sedation while 6121 (87.9 %) received DEX-CH sedation. No significant differences were observed in the sedation success rate with the first dose between the two groups [824 (97.4 %) for DEX vs. 5971 (97.6 %) for DEX-CH; RR 0.99; 95 % CI, 0.98-1.01; P = 0.79]. Similarly, there were no notable disparities in the incidence of adverse events [16 (1.9 %) for DEX vs. 101 (1.7 %) for DEX-CH; RR 1.15; 95 % CI, 0.68-1.93; P = 0.32]. However, intranasal DEX sedation compared with DEX-CH sedation was associated with lower vomiting [0 vs. 95(1.6 %); RR 0.04; 95 % CI, 0.02-0.6; P = 0.02] or more bradycardia [13(1.5 %) vs. 2(0.03 %); RR 47.03; 95 % CI, 10.63-208.04; P < 0.001]. Multivariate analysis using PSM and IPTW analysis yielded similar results. Conclusion Both methods for EEG had high sedation success rate and low incidence of adverse events. High-dose intranasal DEX was more likely to induce bradycardia and had a shorter recovery time than the DEX-CH sedation, which was more likely to induce vomiting.
Collapse
Affiliation(s)
- Liang Wang
- Dazhou Central Hospital, Dazhou 635000, China
| | - Hezhi Wang
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Critical Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Wen Tang
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Critical Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Linlin Tang
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Critical Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Ying Xu
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Critical Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Ling Xiong
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Critical Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| |
Collapse
|
3
|
Beckley JT, Aman TK, Ackley MA, Kazdoba TM, Lewis MC, Smith AC, Farley BJ, Dai J, Deats W, Hoffmann E, Robichaud AJ, Doherty JJ, Quirk MC. Pharmacological characterization of SAGE-718, a novel positive allosteric modulator of N-methyl-d-aspartate receptors. Br J Pharmacol 2024; 181:1028-1050. [PMID: 37698384 DOI: 10.1111/bph.16235] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 07/25/2023] [Accepted: 08/22/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Select neuroactive steroids tune neural activity by modulating excitatory and inhibitory neurotransmission, including the endogenous cholesterol metabolite 24(S)-hydroxycholesterol (24(S)-HC), which is an N-methyl-d-aspartate (NMDA) receptor positive allosteric modulator (PAM). NMDA receptor PAMs are potentially an effective pharmacotherapeutic strategy to treat conditions associated with NMDA receptor hypofunction. EXPERIMENTAL APPROACH Using in vitro and in vivo electrophysiological recording experiments and behavioural approaches, we evaluated the effect of SAGE-718, a novel neuroactive steroid NMDA receptor PAM currently in clinical development for the treatment of cognitive impairment, on NMDA receptor function and endpoints that are altered by NMDA receptor hypoactivity and assessed its safety profile. KEY RESULTS SAGE-718 potentiated GluN1/GluN2A-D NMDA receptors with equipotency and increased NMDA receptor excitatory postsynaptic potential (EPSP) amplitude without affecting decay kinetics in striatal medium spiny neurons. SAGE-718 increased the rate of unblock of the NMDA receptor open channel blocker ketamine on GluN1/GluN2A in vitro and accelerated the rate of return on the ketamine-evoked increase in gamma frequency band power, as measured with electroencephalogram (EEG), suggesting that PAM activity is driven by increased channel open probability. SAGE-718 ameliorated deficits due to NMDA receptor hypofunction, including social deficits induced by subchronic administration of phencyclidine, and behavioural and electrophysiological deficits from cholesterol and 24(S)-HC depletion caused by 7-dehydrocholesterol reductase inhibition. Finally, SAGE-718 did not produce epileptiform activity in a seizure model or neurodegeneration following chronic dosing. CONCLUSIONS AND IMPLICATIONS These findings provide strong evidence that SAGE-718 is a neuroactive steroid NMDA receptor PAM with a mechanism that is well suited as a treatment for conditions associated with NMDA receptor hypofunction.
Collapse
Affiliation(s)
| | - Teresa K Aman
- Sage Therapeutics Inc, Cambridge, Massachusetts, USA
| | | | | | | | - Anne C Smith
- Sage Therapeutics Inc, Cambridge, Massachusetts, USA
| | | | - Jing Dai
- Sage Therapeutics Inc, Cambridge, Massachusetts, USA
| | - Wayne Deats
- Sage Therapeutics Inc, Cambridge, Massachusetts, USA
| | | | | | | | | |
Collapse
|
4
|
Vuu I, Patterson EE, Wu CY, Zolkowska D, Leppik IE, Rogawski MA, Worrell GA, Kremen V, Cloyd JC, Coles LD. Intravenous and Intramuscular Allopregnanolone for Early Treatment of Status Epilepticus: Pharmacokinetics, Pharmacodynamics, and Safety in Dogs. J Pharmacol Exp Ther 2022; 380:104-113. [PMID: 34862270 PMCID: PMC11048262 DOI: 10.1124/jpet.121.000736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/30/2021] [Indexed: 11/22/2022] Open
Abstract
Allopregnanolone (ALLO) is a neurosteroid that modulates synaptic and extrasynaptic GABAA receptors. We hypothesize that ALLO may be useful as first-line treatment of status epilepticus (SE). Our objectives were to (1) characterize ALLO pharmacokinetics-pharmacodynamics PK-PD after intravenous (IV) and intramuscular (IM) administration and (2) compare IV and IM ALLO safety and tolerability. Three healthy dogs and two with a history of epilepsy were used. Single ALLO IV doses ranging from 1-6 mg/kg were infused over 5 minutes or injected IM. Blood samples, vital signs, and sedation assessment were collected up to 8 hours postdose. Intracranial EEG (iEEG) was continuously recorded in one dog. IV ALLO exhibited dose-proportional increases in exposure, which were associated with an increase in absolute power spectral density in all iEEG frequency bands. This relationship was best described by an indirect link PK-PD model where concentration-response was described by a sigmoidal maximum response (Emax) equation. Adverse events included site injection pain with higher IM volumes and ataxia and sedation associated with higher doses. IM administration exhibited incomplete absorption and volume-dependent bioavailability. Robust iEEG changes after IM administration were not observed. Based on PK-PD simulations, a 2 mg/kg dose infused over 5 minutes is predicted to achieve plasma concentrations above the EC50, but below those associated with heavy sedation. This study demonstrates that ALLO is safe and well tolerated when administered at 1-4 mg/kg IV and up to 2 mg/kg IM. The rapid onset of effect after IV infusion suggests that ALLO may be useful in the early treatment of SE. SIGNIFICANCE STATEMENT: The characterization of the pharmacokinetics and pharmacodynamics of allopregnanolone is essential in order to design clinical studies evaluating its effectiveness as an early treatment for status epilepticus in dogs and people. This study has proposed a target dose/therapeutic range for a clinical trial in canine status epilepticus.
Collapse
Affiliation(s)
- Irene Vuu
- Clinical Pharmacology Modeling and Simulation, Amgen Inc., Thousand Oaks, California (I.V.); Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota (E.E.P.); Department of Neurology, University of California Davis School of Medicine, Sacramento, California (C.-Y.W., D.Z., M.A.R.); Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota (I.E.L., J.C.C., L.D.C.); and Department of Neurology, Mayo Clinic, Rochester, Minnesota (G.A.W., V.K.)
| | - Edward E Patterson
- Clinical Pharmacology Modeling and Simulation, Amgen Inc., Thousand Oaks, California (I.V.); Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota (E.E.P.); Department of Neurology, University of California Davis School of Medicine, Sacramento, California (C.-Y.W., D.Z., M.A.R.); Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota (I.E.L., J.C.C., L.D.C.); and Department of Neurology, Mayo Clinic, Rochester, Minnesota (G.A.W., V.K.)
| | - Chun-Yi Wu
- Clinical Pharmacology Modeling and Simulation, Amgen Inc., Thousand Oaks, California (I.V.); Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota (E.E.P.); Department of Neurology, University of California Davis School of Medicine, Sacramento, California (C.-Y.W., D.Z., M.A.R.); Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota (I.E.L., J.C.C., L.D.C.); and Department of Neurology, Mayo Clinic, Rochester, Minnesota (G.A.W., V.K.)
| | - Dorota Zolkowska
- Clinical Pharmacology Modeling and Simulation, Amgen Inc., Thousand Oaks, California (I.V.); Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota (E.E.P.); Department of Neurology, University of California Davis School of Medicine, Sacramento, California (C.-Y.W., D.Z., M.A.R.); Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota (I.E.L., J.C.C., L.D.C.); and Department of Neurology, Mayo Clinic, Rochester, Minnesota (G.A.W., V.K.)
| | - Ilo E Leppik
- Clinical Pharmacology Modeling and Simulation, Amgen Inc., Thousand Oaks, California (I.V.); Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota (E.E.P.); Department of Neurology, University of California Davis School of Medicine, Sacramento, California (C.-Y.W., D.Z., M.A.R.); Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota (I.E.L., J.C.C., L.D.C.); and Department of Neurology, Mayo Clinic, Rochester, Minnesota (G.A.W., V.K.)
| | - Michael A Rogawski
- Clinical Pharmacology Modeling and Simulation, Amgen Inc., Thousand Oaks, California (I.V.); Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota (E.E.P.); Department of Neurology, University of California Davis School of Medicine, Sacramento, California (C.-Y.W., D.Z., M.A.R.); Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota (I.E.L., J.C.C., L.D.C.); and Department of Neurology, Mayo Clinic, Rochester, Minnesota (G.A.W., V.K.)
| | - Gregory A Worrell
- Clinical Pharmacology Modeling and Simulation, Amgen Inc., Thousand Oaks, California (I.V.); Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota (E.E.P.); Department of Neurology, University of California Davis School of Medicine, Sacramento, California (C.-Y.W., D.Z., M.A.R.); Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota (I.E.L., J.C.C., L.D.C.); and Department of Neurology, Mayo Clinic, Rochester, Minnesota (G.A.W., V.K.)
| | - Vaclav Kremen
- Clinical Pharmacology Modeling and Simulation, Amgen Inc., Thousand Oaks, California (I.V.); Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota (E.E.P.); Department of Neurology, University of California Davis School of Medicine, Sacramento, California (C.-Y.W., D.Z., M.A.R.); Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota (I.E.L., J.C.C., L.D.C.); and Department of Neurology, Mayo Clinic, Rochester, Minnesota (G.A.W., V.K.)
| | - James C Cloyd
- Clinical Pharmacology Modeling and Simulation, Amgen Inc., Thousand Oaks, California (I.V.); Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota (E.E.P.); Department of Neurology, University of California Davis School of Medicine, Sacramento, California (C.-Y.W., D.Z., M.A.R.); Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota (I.E.L., J.C.C., L.D.C.); and Department of Neurology, Mayo Clinic, Rochester, Minnesota (G.A.W., V.K.)
| | - Lisa D Coles
- Clinical Pharmacology Modeling and Simulation, Amgen Inc., Thousand Oaks, California (I.V.); Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota (E.E.P.); Department of Neurology, University of California Davis School of Medicine, Sacramento, California (C.-Y.W., D.Z., M.A.R.); Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota (I.E.L., J.C.C., L.D.C.); and Department of Neurology, Mayo Clinic, Rochester, Minnesota (G.A.W., V.K.)
| |
Collapse
|
5
|
Saravanapandian V, Nadkarni D, Hsu SH, Hussain SA, Maski K, Golshani P, Colwell CS, Balasubramanian S, Dixon A, Geschwind DH, Jeste SS. Abnormal sleep physiology in children with 15q11.2-13.1 duplication (Dup15q) syndrome. Mol Autism 2021; 12:54. [PMID: 34344470 PMCID: PMC8336244 DOI: 10.1186/s13229-021-00460-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/21/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Sleep disturbances in autism spectrum disorder (ASD) represent a common and vexing comorbidity. Clinical heterogeneity amongst these warrants studies of the mechanisms associated with specific genetic etiologies. Duplications of 15q11.2-13.1 (Dup15q syndrome) are highly penetrant for neurodevelopmental disorders (NDDs) such as intellectual disability and ASD, as well as sleep disturbances. Genes in the 15q region, particularly UBE3A and a cluster of GABAA receptor genes, are critical for neural development, synaptic protein synthesis and degradation, and inhibitory neurotransmission. During awake electroencephalography (EEG), children with Dup15q syndrome demonstrate increased beta band oscillations (12-30 Hz) that likely reflect aberrant GABAergic neurotransmission. Healthy sleep rhythms, necessary for robust cognitive development, are also highly dependent on GABAergic neurotransmission. We therefore hypothesized that sleep physiology would be abnormal in children with Dup15q syndrome. METHODS To test the hypothesis that elevated beta oscillations persist in sleep in Dup15q syndrome and that NREM sleep rhythms would be disrupted, we computed: (1) beta power, (2) spindle density, and (3) percentage of slow-wave sleep (SWS) in overnight sleep EEG recordings from a cohort of children with Dup15q syndrome (n = 15) and compared them to age-matched neurotypical children (n = 12). RESULTS Children with Dup15q syndrome showed abnormal sleep physiology with elevated beta power, reduced spindle density, and reduced or absent SWS compared to age-matched neurotypical controls. LIMITATIONS This study relied on clinical EEG where sleep staging was not available. However, considering that clinical polysomnograms are challenging to collect in this population, the ability to quantify these biomarkers on clinical EEG-routinely ordered for epilepsy monitoring-opens the door for larger-scale studies. While comparable to other human studies in rare genetic disorders, a larger sample would allow for examination of the role of seizure severity, medications, and developmental age that may impact sleep physiology. CONCLUSIONS We have identified three quantitative EEG biomarkers of sleep disruption in Dup15q syndrome, a genetic condition highly penetrant for ASD. Insights from this study not only promote a greater mechanistic understanding of the pathophysiology defining Dup15q syndrome, but also lay the foundation for studies that investigate the association between sleep and cognition. Abnormal sleep physiology may undermine healthy cognitive development and may serve as a quantifiable and modifiable target for behavioral and pharmacological interventions.
Collapse
Affiliation(s)
- Vidya Saravanapandian
- Center for Autism Research and Treatment, Semel Institute for Neuroscience, University of California, Los Angeles, Los Angeles, CA, 90024, USA. .,Neuroscience Interdepartmental Ph.D. Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Divya Nadkarni
- Division of Pediatric Epilepsy, Department of Pediatric Neurology, Children's Hospital Medical Center of Akron, Akron, OH, 44308, USA
| | - Sheng-Hsiou Hsu
- Swartz Center for Computational Neuroscience, UC San Diego, La Jolla, USA
| | - Shaun A Hussain
- Division of Pediatric Neurology, David Geffen School of Medicine, UCLA Mattel Children's Hospital, Los Angeles, CA, USA
| | - Kiran Maski
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Peyman Golshani
- Department of Neurology and Semel Institute for Neuroscience, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA.,West Los Angeles VA Medical Center, 11301 Wilshire Blvd, Los Angeles, CA, 90073, USA
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | | | - Amos Dixon
- Undergraduate Interdepartmental Program for Neuroscience, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Daniel H Geschwind
- Center for Autism Research and Treatment, Semel Institute for Neuroscience, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | | |
Collapse
|
6
|
Althaus AL, Ackley MA, Belfort GM, Gee SM, Dai J, Nguyen DP, Kazdoba TM, Modgil A, Davies PA, Moss SJ, Salituro FG, Hoffmann E, Hammond RS, Robichaud AJ, Quirk MC, Doherty JJ. Preclinical characterization of zuranolone (SAGE-217), a selective neuroactive steroid GABA A receptor positive allosteric modulator. Neuropharmacology 2020; 181:108333. [PMID: 32976892 PMCID: PMC8265595 DOI: 10.1016/j.neuropharm.2020.108333] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 08/26/2020] [Accepted: 09/18/2020] [Indexed: 01/04/2023]
Abstract
Zuranolone (SAGE-217) is a novel, synthetic, clinical stage neuroactive steroid GABAA receptor positive allosteric modulator designed with the pharmacokinetic properties to support oral daily dosing. In vitro, zuranolone enhanced GABAA receptor current at nine unique human recombinant receptor subtypes, including representative receptors for both synaptic (γ subunit-containing) and extrasynaptic (δ subunit-containing) configurations. At a representative synaptic subunit configuration, α1β2γ2, zuranolone potentiated GABA currents synergistically with the benzodiazepine diazepam, consistent with the non-competitive activity and distinct binding sites of the two classes of compounds at synaptic receptors. In a brain slice preparation, zuranolone produced a sustained increase in GABA currents consistent with metabotropic trafficking of GABAA receptors to the cell surface. In vivo, zuranolone exhibited potent activity, indicating its ability to modulate GABAA receptors in the central nervous system after oral dosing by protecting against chemo-convulsant seizures in a mouse model and enhancing electroencephalogram β-frequency power in rats. Together, these data establish zuranolone as a potent and efficacious neuroactive steroid GABAA receptor positive allosteric modulator with drug-like properties and CNS exposure in preclinical models. Recent clinical data support the therapeutic promise of neuroactive steroid GABAA receptor positive modulators for treating mood disorders; brexanolone is the first therapeutic approved specifically for the treatment of postpartum depression. Zuranolone is currently under clinical investigation for the treatment of major depressive episodes in major depressive disorder, postpartum depression, and bipolar depression.
Collapse
Affiliation(s)
- Alison L Althaus
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA.
| | - Michael A Ackley
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Gabriel M Belfort
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Steven M Gee
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Jing Dai
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - David P Nguyen
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Tatiana M Kazdoba
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Amit Modgil
- Department of Neuroscience, Tufts University, Boston, MA, USA
| | - Paul A Davies
- Department of Neuroscience, Tufts University, Boston, MA, USA
| | - Stephen J Moss
- Department of Neuroscience, Tufts University, Boston, MA, USA
| | - Francesco G Salituro
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Ethan Hoffmann
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Rebecca S Hammond
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Albert J Robichaud
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Michael C Quirk
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - James J Doherty
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| |
Collapse
|
7
|
Abstract
BACKGROUND In 2013 the Food and Drug Administration (FDA) claimed the existence of new data showing women to be at risk for excessive daytime sedation and impaired driving proficiency following bedtime doses of zolpidem. The putative explanation was the reduced metabolic clearance of zolpidem and higher morning blood concentrations in women compared to men. The FDA acted to reduce the recommended dosage for women down to 50% of the dose for men. No other regulatory agency worldwide has taken similar action. METHODS Gender effects on zolpidem pharmacokinetics, pharmacodynamics, adverse effects, clinical efficacy, and driving performance were evaluated through a further analysis of data from a previous study, together with a literature review. RESULTS Women had on average 35% lower apparent clearance of zolpidem than men (236 vs 364 mL/min, P < 0.001). This difference was not explained by body weight. In some laboratory studies, women had greater functional impairment than men taking the same dose, but in all studies active drug was not distinguishable from placebo at 8 hours after oral dosage. On-the-road driving studies likewise showed no evidence of driving impairment in men or women at 8 hours after 10 mg of oral immediate-release zolpidem. No clinical trial demonstrated a gender-related difference in clinical efficacy or adverse reactions, and there was no evidence of a particular risk to women. CONCLUSIONS Dosage reduction in women is not supported by available scientific evidence, and may in fact lead to underdosing and the consequent hazard of inadequately treated insomnia.
Collapse
|
8
|
Frohlich J, Reiter LT, Saravanapandian V, DiStefano C, Huberty S, Hyde C, Chamberlain S, Bearden CE, Golshani P, Irimia A, Olsen RW, Hipp JF, Jeste SS. Mechanisms underlying the EEG biomarker in Dup15q syndrome. Mol Autism 2019; 10:29. [PMID: 31312421 PMCID: PMC6609401 DOI: 10.1186/s13229-019-0280-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
Background Duplications of 15q11.2-q13.1 (Dup15q syndrome), including the paternally imprinted gene UBE3A and three nonimprinted gamma-aminobutyric acid type-A (GABAA) receptor genes, are highly penetrant for neurodevelopmental disorders such as autism spectrum disorder (ASD). To guide targeted treatments of Dup15q syndrome and other forms of ASD, biomarkers are needed that reflect molecular mechanisms of pathology. We recently described a beta EEG phenotype of Dup15q syndrome, but it remains unknown which specific genes drive this phenotype. Methods To test the hypothesis that UBE3A overexpression is not necessary for the beta EEG phenotype, we compared EEG from a reference cohort of children with Dup15q syndrome (n = 27) to (1) the pharmacological effects of the GABAA modulator midazolam (n = 12) on EEG from healthy adults, (2) EEG from typically developing (TD) children (n = 14), and (3) EEG from two children with duplications of paternal 15q (i.e., the UBE3A-silenced allele). Results Peak beta power was significantly increased in the reference cohort relative to TD controls. Midazolam administration recapitulated the beta EEG phenotype in healthy adults with a similar peak frequency in central channels (f = 23.0 Hz) as Dup15q syndrome (f = 23.1 Hz). Both paternal Dup15q syndrome cases displayed beta power comparable to the reference cohort. Conclusions Our results suggest a critical role for GABAergic transmission in the Dup15q syndrome beta EEG phenotype, which cannot be explained by UBE3A dysfunction alone. If this mechanism is confirmed, the phenotype may be used as a marker of GABAergic pathology in clinical trials for Dup15q syndrome.
Collapse
Affiliation(s)
- Joel Frohlich
- Roche Pharma Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
- Center for Autism Research and Treatment, University of California Los Angeles, Semel Institute for Neuroscience, Los Angeles, CA 90024 USA
- Department of Psychology, University of California Los Angeles, 3423 Franz Hall, Los Angeles, CA 90095 USA
| | - Lawrence T. Reiter
- Departments of Neurology, Pediatrics and Anatomy & Neurobiology, The University of Tennessee Health Science Center, 855 Monroe Ave., Link, Memphis, TN 415 USA
| | - Vidya Saravanapandian
- Center for Autism Research and Treatment, University of California Los Angeles, Semel Institute for Neuroscience, Los Angeles, CA 90024 USA
| | - Charlotte DiStefano
- Center for Autism Research and Treatment, University of California Los Angeles, Semel Institute for Neuroscience, Los Angeles, CA 90024 USA
| | - Scott Huberty
- Center for Autism Research and Treatment, University of California Los Angeles, Semel Institute for Neuroscience, Los Angeles, CA 90024 USA
- McGill University, MUHC Research Institute, 5252, boul. de Maisonneuve Ouest, 3E.19, Montreal, QC H4A 3S5 Canada
| | - Carly Hyde
- Center for Autism Research and Treatment, University of California Los Angeles, Semel Institute for Neuroscience, Los Angeles, CA 90024 USA
| | - Stormy Chamberlain
- Genetics and Genome Sciences, UConn Health, 400 Farmington Avenue, Farmington, CT 06030-6403 USA
| | - Carrie E. Bearden
- Department of Psychiatry and Biobehavioral Sciences and Department of Psychology, University of California Los Angeles, Suite A7-460, 760 Westwood Plaza, Los Angeles, CA 90095 USA
| | - Peyman Golshani
- Department of Neurology and Psychiatry, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA 90095 USA
| | - Andrei Irimia
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave., Suite 228C, California, Los Angeles 90089 USA
| | - Richard W. Olsen
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, California, Los Angeles 90095 USA
| | - Joerg F. Hipp
- Roche Pharma Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
| | - Shafali S. Jeste
- Center for Autism Research and Treatment, University of California Los Angeles, Semel Institute for Neuroscience, Los Angeles, CA 90024 USA
| |
Collapse
|
9
|
Pagel JF, Pandi-Perumal SR, Monti JM. Treating insomnia with medications. SLEEP SCIENCE AND PRACTICE 2018. [DOI: 10.1186/s41606-018-0025-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
10
|
Chen X, Broeyer F, de Kam M, Baas J, Cohen A, van Gerven J. Pharmacodynamic response profiles of anxiolytic and sedative drugs. Br J Clin Pharmacol 2017; 83:1028-1038. [PMID: 27922194 DOI: 10.1111/bcp.13204] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 02/04/2023] Open
Abstract
AIM Centrally-acting acutely anxiolytic drugs, such as benzodiazepines, barbiturates and gabapentinoids, affect various central nervous system (CNS) functions, which reflects not only their anxiolytic effects but also neuropsychological side-effects. To validate the pharmacodynamic biomarkers for GABA-ergic anxiolytics, this study determined the pharmacodynamics of two anxiolytics and a nonanxiolytic control, and linked them to their anxiolytic and sedative effects, during an anxiety-challenge study day. METHODS Twenty healthy volunteers were randomized in this placebo-controlled, double-blind, four-way cross-over study with single-dose alprazolam (1 mg), diphenhydramine (50 mg), pregabalin (200 mg) or placebo. The Neurocart was used between repeated fear-potentiated startle assessments. Thus, the potential influence of anxiety on CNS pharmacodynamic markers could be examined. RESULTS Compared to placebo, VAScalmness increased with alprazolam (2.0 mm) and pregabalin (2.5 mm) but not with diphenhydramine. Saccadic peak velocity (SPV) declined after alprazolam (-57 ° s-1 ) and pregabalin (-28 ° s-1 ), more than with diphenhydramine (-14 ° s-1 ); so did smooth pursuit. The average responses of SPV and smooth pursuit were significantly correlated with the drug-induced increases in VAScalmness . The SPV-relative responses of VASalertness , body-sway and adaptive-tracking also differed among alprazolam, pregabalin and diphenhydramine. CONCLUSIONS Compared with the antihistaminergic sedative diphenhydramine, alprazolam and pregabalin caused larger SPV reduction, which was correlated with simultaneous improvement of subjective calmness, during a study day in which anxiety was stimulated repeatedly. The different effect profiles of the three drugs are in line with their pharmacological distinctions. These findings corroborate the profiling of CNS effects to demonstrate pharmacological selectivity, and further support SPV as biomarker for anxiolysis involving GABA-ergic neurons. The study also supports the use of prolonged mild threat to demonstrate anxiolytic effects in healthy volunteers.
Collapse
Affiliation(s)
- Xia Chen
- Phase I Unit, Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Beijing, 100032, China.,Centre for Human Drug Research, Leiden, The Netherlands
| | | | | | - Joke Baas
- Department of Experimental Psychology, Faculty of Social Sciences, Utrecht University, The Netherlands
| | - Adam Cohen
- Centre for Human Drug Research, Leiden, The Netherlands
| | | |
Collapse
|
11
|
Hill L, Bertaccini E, Barr J, Geller E. ICU Sedation: A Review of Its Pharmacology and Assessment. J Intensive Care Med 2016. [DOI: 10.1177/088506669801300405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The need for appropriate sedation in the intensive care unit is paramount. Critically ill patients are exposed to multiple adverse stimuli stemming from both their illness and their environment. If left unchecked, these stimuli may often produce potentially harmful physiologic sequelae in patients who already have compromised physiologic reserve. The most useful sedative agents in such circumstances are those which are readily titratable and have manageable side effects. This typically focuses discussion on the intravenous administration of analgesic sedatives (opioids), anxiolytic and amnestic sedatives (benzodiazepines, barbiturates, etomidate, propofol), dissociative sedatives (ketamine), and the antipsychotic sedatives (butyrophenones). With ready titratability, though, comes the need for efficient monitoring and assessment of the degree of sedation. While no measure is without bias, this can effectively be done via the subjective means of a sedation scoring scheme or the more objective means of electrophysiologic measurements. It is the combination of pharmacological tools and consistent assessment which will allow the intensivist to readily achieve the desired sedation goal.
Collapse
Affiliation(s)
- Laureen Hill
- Stanford University School of Medicine, Stanford, CA
| | - Ed Bertaccini
- Stanford University School of Medicine, Stanford, CA
| | - Juli Barr
- Stanford University School of Medicine, Stanford, CA, Stanford University School of Medicine, Stanford, CA
| | - Eran Geller
- Stanford University School of Medicine, Stanford, CA, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
12
|
Wilson FJ, Danjou P. Early Decision-Making in Drug Development: The Potential Role of Pharmaco-EEG and Pharmaco-Sleep. Neuropsychobiology 2016; 72:188-94. [PMID: 26900685 DOI: 10.1159/000382022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/30/2015] [Indexed: 11/19/2022]
Abstract
The pharmaceutical industry has been suffering from low clinical success rates of new drugs for some time with particularly high attrition in early clinical development, especially for drugs aimed at central targets. Both pharmaco-electroencephalography (EEG) and pharmaco-sleep, along with other biomarker techniques, have significant potential to assist with this problem by enabling early decisions to be made about the likelihood of a compound proving successful in the clinic. This paper discusses the role and points of application of biomarker techniques in early drug development. It proposes a framework for the use of pharmaco-EEG and pharmaco-sleep in drug development that (i) relies on the combination of preclinical data and an understanding of translatability to generate robust hypotheses for testing in early clinical studies and (ii) is backed up by a clear decision-making process. The areas that need further development before this framework can be put fully into practice are discussed, along with some possible routes by which this could be achieved through precompetitive co-operation within the industry.
Collapse
|
13
|
Drinkenburg WHIM, Ahnaou A, Ruigt GSF. Pharmaco-EEG Studies in Animals: A History-Based Introduction to Contemporary Translational Applications. Neuropsychobiology 2016; 72:139-50. [PMID: 26901675 DOI: 10.1159/000443175] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Current research on the effects of pharmacological agents on human neurophysiology finds its roots in animal research, which is also reflected in contemporary animal pharmaco-electroencephalography (p-EEG) applications. The contributions, present value and translational appreciation of animal p-EEG-based applications are strongly interlinked with progress in recording and neuroscience analysis methodology. After the pioneering years in the late 19th and early 20th century, animal p-EEG research flourished in the pharmaceutical industry in the early 1980s. However, around the turn of the millennium the emergence of structurally and functionally revealing imaging techniques and the increasing application of molecular biology caused a temporary reduction in the use of EEG as a window into the brain for the prediction of drug efficacy. Today, animal p-EEG is applied again for its biomarker potential - extensive databases of p-EEG and polysomnography studies in rats and mice hold EEG signatures of a broad collection of psychoactive reference and test compounds. A multitude of functional EEG measures has been investigated, ranging from simple spectral power and sleep-wake parameters to advanced neuronal connectivity and plasticity parameters. Compared to clinical p-EEG studies, where the level of vigilance can be well controlled, changes in sleep-waking behaviour are generally a prominent confounding variable in animal p-EEG studies and need to be dealt with. Contributions of rodent pharmaco-sleep EEG research are outlined to illustrate the value and limitations of such preclinical p-EEG data for pharmacodynamic and chronopharmacological drug profiling. Contemporary applications of p-EEG and pharmaco-sleep EEG recordings in animals provide a common and relatively inexpensive window into the functional brain early in the preclinical and clinical development of psychoactive drugs in comparison to other brain imaging techniques. They provide information on the impact of drugs on arousal and sleep architecture, assessing their neuropharmacological characteristics in vivo, including central exposure and information on kinetics. In view of the clear disadvantages as well as advantages of animal p-EEG as compared to clinical p-EEG, general statements about the usefulness of EEG as a biomarker to demonstrate the translatability of p-EEG effects should be made with caution, however, because they depend on the particular EEG or sleep parameter that is being studied. The contribution of animal p-EEG studies to the translational characterisation of centrally active drugs can be furthered by adherence to guidelines for methodological standardisation, which are presently under construction by the International Pharmaco-EEG Society (IPEG).
Collapse
|
14
|
Sedation for electroencephalography with dexmedetomidine or chloral hydrate: a comparative study on the qualitative and quantitative electroencephalogram pattern. J Neurosurg Anesthesiol 2016; 27:21-5. [PMID: 24823763 DOI: 10.1097/ana.0000000000000077] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Sedation for electroencephalography in uncooperative patients is a controversial issue because majority of sedatives, hypnotics, and general anesthetics interfere with the brain's electrical activity. Chloral hydrate (CH) is typically used for this sedation, and dexmedetomidine (DEX) was recently tested because preliminary data suggest that this drug does not affect the electroencephalogram (EEG). The aim of the present study was to compare the EEG pattern during DEX or CH sedation to test the hypothesis that both drugs exert similar effects on the EEG. MATERIALS AND METHODS A total of 17 patients underwent 2 EEGs on 2 separate occasions, one with DEX and the other with CH. The EEG qualitative variables included the phases of sleep and the background activity. The EEG quantitative analysis was performed during the first 2 minutes of the second stage of sleep. The EEG quantitative variables included density, duration, and amplitude of the sleep spindles and absolute spectral power. RESULTS The results showed that the qualitative analysis, density, duration, and amplitude of sleep spindles did not differ between DEX and CH sedation. The power of the slow-frequency bands (δ and θ) was higher with DEX, but the power of the faster-frequency bands (α and β) was higher with CH. The total power was lower with DEX than with CH. CONCLUSIONS The differences of DEX and CH in EEG power did not change the EEG qualitative interpretation, which was similar with the 2 drugs. Other studies comparing natural sleep and sleep induced by these drugs are needed to clarify the clinical relevance of the observed EEG quantitative differences.
Collapse
|
15
|
GABA withdrawal syndrome: GABAA receptor, synapse, neurobiological implications and analogies with other abstinences. Neuroscience 2015; 313:57-72. [PMID: 26592722 DOI: 10.1016/j.neuroscience.2015.11.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 11/07/2015] [Accepted: 11/10/2015] [Indexed: 11/22/2022]
Abstract
The sudden interruption of the increase of the concentration of the gamma-aminobutyric acid (GABA), determines an increase in neuronal activity. GABA withdrawal (GW) is a heuristic analogy, with withdrawal symptoms developed by other GABA receptor-agonists such as alcohol, benzodiazepines, and neurosteroids. GW comprises a model of neuronal excitability validated by electroencephalogram (EEG) in which high-frequency and high-amplitude spike-wave complexes appear. In brain slices, GW was identified by increased firing synchronization of pyramidal neurons and by changes in the active properties of the neuronal membrane. GW induces pre- and postsynaptic changes: a decrease in GABA synthesis/release, and the decrease in the expression and composition of GABAA receptors associated with increased calcium entry into the cell. GW is an excellent bioassay for studying partial epilepsy, epilepsy refractory to drug treatment, and a model to reverse or prevent the generation of abstinences from different drugs.
Collapse
|
16
|
Jones KA, Menniti FS, Sivarao DV. Translational psychiatry-light at the end of the tunnel. Ann N Y Acad Sci 2015; 1344:1-11. [DOI: 10.1111/nyas.12725] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
17
|
Salansky N, Fedotchev A, Bondar A. High-Frequency Resolution EEG: Results and Opportunities. ACTA ACUST UNITED AC 2015. [DOI: 10.1080/00029238.1995.11080508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Norman Salansky
- Institute for Aerospace Studies, University of Toronto, Downsview, Ontario, M3H 5T6, Canada
| | - Alexander Fedotchev
- International Medical Instruments, Inc., 1520 Steeles Ave. W., Concord, Ontario, L4K 3B9, Canada
| | - Alexander Bondar
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino Moscow Region, 142292, Russia
| |
Collapse
|
18
|
Keidan I, Ben-Menachem E, Tzadok M, Ben-Zeev B, Berkenstadt H. Electroencephalography for children with autistic spectrum disorder: a sedation protocol. Paediatr Anaesth 2015; 25:200-5. [PMID: 25145661 DOI: 10.1111/pan.12510] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/23/2014] [Indexed: 12/25/2022]
Abstract
OBJECTIVES To report the effectiveness and efficiency of a predetermined sedation protocol for providing sedation for electroencephalograph (EEG) studies in children with autism. METHODS Sleep EEG has been advocated for the majority of children with autism spectrum disorder. In most cases, sedation is required to allow adequate studies. Most sedation drugs have negative effects on the EEG pattern. The sedation protocol we adopted included chloral hydrate, dexmedetomidine, and ketamine and was evaluated prospectively for 2 years. RESULTS One hundred and eighty-three children with autistic spectrum disorder were sedated with the described drug protocol that was efficient, provided adequate EEG readings, and was not associated with serious adverse events. CONCLUSIONS Our protocol kept costs to a minimum but provided appropriate escalation in care when required.
Collapse
Affiliation(s)
- Ilan Keidan
- Department of Anesthesiology and Intensive Care, Sheba Medical Center, Tel Hashomer, Israel; Department of Anesthesiology, University of Florida, Gainesville, FL, USA
| | | | | | | | | |
Collapse
|
19
|
|
20
|
English BA, Thomas K, Johnstone J, Bazih A, Gertsik L, Ereshefsky L. Use of translational pharmacodynamic biomarkers in early-phase clinical studies for schizophrenia. Biomark Med 2014; 8:29-49. [PMID: 24325223 DOI: 10.2217/bmm.13.135] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Schizophrenia is a severe mental disorder characterized by cognitive deficits, and positive and negative symptoms. The development of effective pharmacological compounds for the treatment of schizophrenia has proven challenging and costly, with many compounds failing during clinical trials. Many failures occur due to disease heterogeneity and lack of predictive preclinical models and biomarkers that readily translate to humans during early characterization of novel antipsychotic compounds. Traditional early-phase trials consist of single- or multiple-dose designs aimed at determining the safety and tolerability of an investigational compound in healthy volunteers. However, by incorporating a translational approach employing methodologies derived from preclinical studies, such as EEG measures and imaging, into the traditional Phase I program, critical information regarding a compound's dose-response effects on pharmacodynamic biomarkers can be acquired. Furthermore, combined with the use of patients with stable schizophrenia in early-phase clinical trials, significant 'de-risking' and more confident 'go/no-go' decisions are possible.
Collapse
|
21
|
|
22
|
PKPD Aspects of Brain Drug Delivery in a Translational Perspective. DRUG DELIVERY TO THE BRAIN 2014. [DOI: 10.1007/978-1-4614-9105-7_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
23
|
Thompson C, Shabanova V, Giuliano JS. The SNAP index does not correlate with the State Behavioral Scale in intubated and sedated children. Paediatr Anaesth 2013; 23:1174-9. [PMID: 24103039 PMCID: PMC3880626 DOI: 10.1111/pan.12258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/09/2013] [Indexed: 11/27/2022]
Abstract
BACKGROUND Ensuring appropriate levels of sedation for critically ill children is integral to pediatric critical care. Traditionally, clinicians have used subjective scoring tools to assess sedation levels. The SNAP II uses dual frequency processed electroencephalography to evaluate brain activity and may provide an objective assessment of sedation levels. OBJECTIVE This study attempts to find an objective method to monitor sedation in critically ill pediatric patients. We compared the SNAP II, a processed electroencephalography device, with the State Behavioral Scale (SBS), a subjective sedation scoring tool. We hypothesize that the SNAP II correlates with the SBS and has less observer bias. METHODS This was an IRB approved prospective, observational study. Patients receiving intravenous sedation while being mechanically ventilated were enrolled after informed consent. After the SNAP II monitoring electrodes were attached, blinded bedside nurses assessed sedation levels using the SBS. SNAP indices were collected and compared with SBS scores to determine correlation. RESULTS We compared 417 paired data points from 15 patients using Pearson's correlation and least squares means to determine correlation between the SBS and SNAP indices. No correlation was observed. Using covariance model patterning for repeated measures to adjust for covariates again showed no correlation. CONCLUSION The SNAP index does not correlate with SBS scores in our pediatric intensive care unit (PICU). Its use cannot be recommended to measure levels of sedation in our population. Future research should continue to explore objective ways of measuring sedation in critically ill children.
Collapse
Affiliation(s)
- Cecilia Thompson
- Department of Pediatrics/Division of Critical Care Medicine/Icahn School of Medicine at Mount Sinai/New York/USA
| | | | - John S. Giuliano
- Department of Pediatrics/Division of Critical Care Medicine/Yale University School of Medicine/New Haven/USA
| |
Collapse
|
24
|
Conrado DJ, Bewernitz M, Ding M, Cibula J, Seubert C, Sy SKB, Eisenschenk S, Derendorf H. Electroencephalogram effects of armodafinil: comparison with behavioral alertness. J Clin Pharmacol 2013; 53:1058-71. [PMID: 23913585 DOI: 10.1002/jcph.143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/27/2013] [Indexed: 11/06/2022]
Abstract
Development of central nervous system-acting drugs would be enhanced by suitable biomarkers that reflect the targeted pathophysiologic brain state. The electroencephalogram (EEG) has several characteristics of an ideal biomarker and can be promptly adapted to pre-clinical and clinical testing. The aim of this study was to evaluate EEG as a measure of the wakefulness-promoting effect of armodafinil in sleep deprived healthy subjects. Armodafinil pharmacodynamics were simultaneously assessed by EEG- and behavioral-based measures including a well-established measure of alertness. Using two quantitative EEG-based measures-power spectral and event-related brain activity analyses-we observed that armodafinil mitigated the slowing of brain activity and the decrease of the event-related brain activity caused by sleep deprivation. Armodafinil-induced changes in EEG are in agreement and explain up to 73.1% of the armodafinil-induced changes in alertness. Our findings suggest that EEG can serve as a marker of the wakefulness-promoting drug effect.
Collapse
Affiliation(s)
- Daniela J Conrado
- Department of Pharmaceutics, University of Florida, Gainesville, FL, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Greenblatt DJ, Legangneux E, Harmatz JS, Weinling E, Freeman J, Rice K, Zammit GK. Dynamics and Kinetics of a Modified-Release Formulation of Zolpidem: Comparison With Immediate-Release Standard Zolpidem and Placebo. J Clin Pharmacol 2013; 46:1469-80. [PMID: 17101746 DOI: 10.1177/0091270006293303] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Modified-release (MR) zolpidem was developed to maintain effective plasma concentrations during the 3- to 6-hour post-dosage interval, corresponding to the middle portion of the typical sleep interval. Modified-release zolpidem (12.5 mg), standard immediate-release (IR) zolpidem (10 mg), and placebo were compared in a double-blind, single-dose, 3-way crossover daytime study of healthy volunteers (n = 70 completers). Effect areas for electroencephalographic beta amplitude during 0 to 8 hours and 3 to 6 hours after dosage were greater for MR compared to IR (P < .001). The digit-symbol substitution test and sedation rating scales behaved similarly. MR and IR did not differ in effects at 8 hours post-dosage nor in halflife or clearance. Time of peak plasma concentration (tmax) was significantly longer for MR (2.4 vs 2.0 hours, P < .004), and dose-normalized peak plasma concentration (Cmax) was lower (12.2 vs 14.0 ng/mL/mg, P < .001). MR zolpidem also had greater area under the plasma concentration curve (AUC) during the 3- to 6-hour interval (P < .001). Thus, MR zolpidem produces sustained plasma levels compared to IR, with resulting enhancement of pharmacodynamic effects in the 3- to 6-hour post-dosage interval.
Collapse
Affiliation(s)
- David J Greenblatt
- Department of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA.
| | | | | | | | | | | | | |
Collapse
|
26
|
Orexin Receptor Antagonism, a New Sleep-Promoting Paradigm: An Ascending Single-Dose Study With Almorexant. Clin Pharmacol Ther 2010; 87:593-600. [DOI: 10.1038/clpt.2010.19] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
27
|
New insights into neuromodulatory approaches for the treatment of pain. THE JOURNAL OF PAIN 2007; 9:193-9. [PMID: 18096437 DOI: 10.1016/j.jpain.2007.11.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Revised: 10/23/2007] [Accepted: 11/01/2007] [Indexed: 11/22/2022]
Abstract
UNLABELLED Two lines of evidence about the association between the experience of pain and brain state (measured via electroencephalogram or EEG) have recently come to light. First, research from a number of sources suggests a link between brain EEG activity and the experience of pain. Specifically, this research suggests that the subjective experience of pain is associated with relatively lower amplitudes of slower wave (delta, theta, and alpha) activity and relatively higher amplitudes of faster wave (beta) activity. Second, there has been a recent increase in interest in interventions that impact the cortical neuromodulation of pain, including behavioral treatments (such as self-hypnosis training and neurofeedback) and both invasive and noninvasive brain stimulation. Although a direct causal link between experience of pain and brain activity as measured by EEG has not been established, the targeting of pain treatment at a cortical level by trying to affect EEG rhythms directly is an intriguing possibility. PERSPECTIVE Preliminary evidence suggests the possibility, which has not yet adequately tested or proven, that the experience of chronic pain is linked to cortical activity as assessed via an electroencephalogram. Support for this hypothesis would have important implications for understanding the mechanisms that underlie a number of pain treatments, and for developing new innovative treatments for chronic pain management.
Collapse
|
28
|
Liefaard LC, Gunput RAF, Danhof M, Voskuyl RA. Decreased Efficacy of GABAA-receptor Modulation by Midazolam in the Kainate Model of Temporal Lobe Epilepsy. Epilepsia 2007; 48:1378-87. [PMID: 17442008 DOI: 10.1111/j.1528-1167.2007.01081.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE The objective of this investigation was to characterize quantitatively the time-dependent changes in midazolam (MDL) efficacy in the silent period after induction of status epilepticus (SE) in rats. The changes in MDL efficacy were correlated to changes in ex vivo GABA(A)-receptor expression. METHODS MDL efficacy was quantified by pharmacokinetic-pharmacodynamic (PK-PD) modeling by using the beta-frequency of the EEG as PD end point. Two PK-PD experiments were performed in each animal: the first experiment before and the second experiment at either day 4 or day 14 after SE. SE was induced by repetitive intraperitoneal injections with kainate. GABA(A)-receptor expression was determined by ex vivo autoradiography with [(3)H]flumazenil. RESULTS The concentration versus EEG effect relation of midazolam was successfully described by the sigmoidal E(max) model. The maximal effect on the beta-frequency of the EEG (E(max)) was reduced to 51.6 +/- 35.6% and 25.8 +/- 33.7% of the original value at 4 and 14 days after induction of SE. The ex vivo study with [(3)H]flumazenil showed that the observed reductions in E(max) were paralleled by a reduction in GABA(A)-receptor density. CONCLUSIONS The efficacy of MDL is decreased in the silent period after SE, which can be partly accounted for by a reduction in GABA(A)-receptor density.
Collapse
Affiliation(s)
- Lia C Liefaard
- Division of Pharmacology, LACDR, Leiden University, Leiden, The Netherlands
| | | | | | | |
Collapse
|
29
|
Mager DE, Abernethy DR. Use of wavelet and fast Fourier transforms in pharmacodynamics. J Pharmacol Exp Ther 2007; 321:423-30. [PMID: 17142645 DOI: 10.1124/jpet.106.113183] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Progress has been made in the development and application of mechanism-based pharmacodynamic models for describing the drug-specific and physiological factors influencing the time course of responses to the diverse actions of drugs. However, the biological variability in biosignals and the complexity of pharmacological systems often complicate or preclude the direct application of traditional structural and nonstructural models. Mathematical transforms may be used to provide measures of drug effects, identify structural and temporal patterns, and visualize multidimensional data from analyses of biomedical signals and images. Fast Fourier transform (FFT) and wavelet analyses are two methodologies that have proven to be useful in this context. FFT converts a signal from the time domain to the frequency domain, whereas wavelet transforms colocalize in both domains and may be utilized effectively for nonstationary signals. Nonstationary drug effects are common but have not been well analyzed and characterized by other methods. In this review, we discuss specific applications of these transforms in pharmacodynamics and their potential role in ascertaining the dynamics of spatiotemporal properties of complex pharmacological systems.
Collapse
Affiliation(s)
- Donald E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, the State Universitiy of New York, Buffalo, NY, USA
| | | |
Collapse
|
30
|
Gilles C, Luthringer R. Pharmacological models in healthy volunteers: their use in the clinical development of psychotropic drugs. J Psychopharmacol 2007; 21:272-82. [PMID: 17591655 DOI: 10.1177/0269881107077733] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Animal models of diseases are widely used in the preclinical phase of drug development. They have a place in early human clinical psychopharmacology as well, in order to get early clues that contribute to establish the proof of concept (POC) already in healthy volunteers (HV). Different types of models are available (pharmacological or non-pharmacological provocation, models based on age-related characteristics). This review is focused on pharmacological models in HV, with the aim to identify the main issues raised by their use in pharmaceutical trials. The available models unevenly fulfil the requirements of face validity, sufficient response rate, test-retest consistence and responsiveness to reference drugs. Most of them have been developed in the purpose of pathophysiology studies, using rating instruments validated for clinical practice. Substantial progress could be made by adapting models to the specific requirements of pharmaceutical trials, including wider use of biomarkers. Characteristics that make models, as well as biomarkers, suitabLe for use in drug development are proposed. Despite obvious limitations, human models can significantly enhance the way phase I studies contribute to establish the POC, provided they are integrated into adapted phase I development plans. Their use as industrial tools for drug evaluation requires specific, dedicated development.
Collapse
|
31
|
Danhof M, de Jongh J, De Lange ECM, Della Pasqua O, Ploeger BA, Voskuyl RA. Mechanism-Based Pharmacokinetic-Pharmacodynamic Modeling: Biophase Distribution, Receptor Theory, and Dynamical Systems Analysis. Annu Rev Pharmacol Toxicol 2007; 47:357-400. [PMID: 17067280 DOI: 10.1146/annurev.pharmtox.47.120505.105154] [Citation(s) in RCA: 174] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mechanism-based PK-PD models differ from conventional PK-PD models in that they contain specific expressions to characterize, in a quantitative manner, processes on the causal path between drug administration and effect. This includes target site distribution, target binding and activation, pharmacodynamic interactions, transduction, and homeostatic feedback mechanisms. As the final step, the effects on disease processes and disease progression are considered. Particularly through the incorporation of concepts from receptor theory and dynamical systems analysis, important progress has been made in the field of mechanism-based PK-PD modeling. This has yielded models with much-improved properties for extrapolation and prediction. These models constitute a theoretical basis for rational drug discovery and development.
Collapse
Affiliation(s)
- Meindert Danhof
- Leiden/Amsterdam Center for Drug Research, Division of Pharmacology, Leiden University, 2300 RA Leiden, The Netherlands.
| | | | | | | | | | | |
Collapse
|
32
|
Barbanoj MJ, Antonijoan RM, Riba J, Valle M, Romero S, Jané F. Quantifying drug-drug interactions in pharmaco-EEG. Clin EEG Neurosci 2006; 37:108-20. [PMID: 16733942 DOI: 10.1177/155005940603700208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A drug interaction refers to an event in which the usual pharmacological effect of a drug is modified by other factors, most frequently additional drugs. When two drugs are administered simultaneously, or within a short time of each other, an interaction can occur that may increase or decrease the intended magnitude or duration of the effect of one or both drugs. Drugs may interact on a pharmaceutical, pharmacokinetic or pharmacodynamic basis. Pharmacodynamic interactions arise when the alteration of the effects occurs at the site of action. This is a wide field where not only interactions between different drugs are considered but also drug and metabolites (midazolam/alpha-hydroxy-midazolam), enantiomers (ketamine), as well as phenomena such as tolerance (nordiazepam) and sensitization (diazepam). Pharmacodynamic interactions can result in antagonism or synergism and can originate at a receptor level (antagonism, partial agonism, down-regulation, up-regulation), at an intraneuronal level (transduction, uptake), or at an interneuronal level (physiological pathways). Alternatively, psychotropic drug interactions assessed through quantitative pharmaco-EEG can be viewed according to the broad underlying objective of the study: safety-oriented (ketoprofen/theophylline, lorazepam/diphenhydramine, granisetron/haloperidol), strictly pharmacologically-oriented (benzodiazepine receptors), or broadly neuro-physiologically-oriented (diazepam/buspirone). Methodological issues are stressed, particularly drug plasma concentrations, dose-response relationships and time-course of effects (fluoxetine/buspirone), and unsolved questions are addressed (yohimbine/caffeine, hydroxizyne/alcohol).
Collapse
Affiliation(s)
- M J Barbanoj
- Centre d'lnvestigació de Medicaments, Institut de Recerca, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau.
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
The main topic of this overview is an analysis of the concepts of phase and synchrony, as used in neurophysiology, in their various meanings. A number of notions related to the concepts of phase and synchrony, which are incorporated in contemporary neurophysiology, particularly in the domain of neuro-cognitive physiology are discussed. These notions need a critical examination, since their use sometimes is not clear, or it may even be ambiguous. We present some of these concepts, namely (a) (des)synchronization, (b) phase resetting, (c) phase synchrony and phase/time delays, and (d) phase clustering within one signal, while discussing what type of neuronal activities may underlie these EEG phenomena.
Collapse
Affiliation(s)
- Fernando H Lopes da Silva
- Center of Neurosciences, Swammerdam Institute for Life Sciences, University of Amsterdam, Kruislaan 320, 1098 SM Amsterdam, The Netherlands.
| |
Collapse
|
34
|
Abstract
Only a few years ago, if patients complained of difficulty sleeping,medications that were often dangerous and addictive were prescribed to induce sleep, and the basis of the patient's complaint was not addressed. Now sleeping pills are safer, and our understanding of the sleep state has increased exponentially. Insomnia and daytime sleepiness are no longer diagnoses; they are complaints needing to be addressed-symptoms of a spectrum of sleep disorders with specific diagnostic criteria and appropriate treatments.
Collapse
Affiliation(s)
- J F Pagel
- Department of Family Practice, University of Colorado School of Medicine, PO Box 6508, Aurora, CO 80045-0508, USA.
| |
Collapse
|
35
|
Greenblatt DJ, Gan L, Harmatz JS, Shader RI. Pharmocokinetics and pharmacodynamics of single-dose triazolam: electroencephalography compared with the Digit-Symbol Substitution Test. Br J Clin Pharmacol 2005; 60:244-8. [PMID: 16120062 PMCID: PMC1884767 DOI: 10.1111/j.1365-2125.2005.02409.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2004] [Accepted: 12/28/2004] [Indexed: 01/22/2023] Open
Abstract
AIMS To investigate whether the electroencephalogram (EEG) directly reflects the CNS effects of benzodiazepines by evaluating the relation of the EEG to plasma drug concentrations and to Digit-Symbol Substitution Test (DSST) scores after a single dose of triazolam, a representative benzodiazepine agonist. METHODS Thirteen healthy male subjects were given 0.375 mg triazolam or placebo in a double-blind crossover study. Plasma samples were collected during 8 h after dosage. Pharmacodynamic effects were measured by DSST and EEG at corresponding times. RESULTS Pharmacokinetic parameters for triazolam were consistent with established values. Compared with placebo, triazolam significantly impaired psychomotor performance on the DSST (P < 0.001) and increased beta amplitude on the EEG (P < 0.002). DSST and EEG changes both closely tracked changes in plasma concentrations over time. The changes for the two measures were highly correlated with each other (r =-0.94, P < 0.001) based on aggregate values at individual time points. However, the variations in area under the curve of pharmacodynamic effect vs. time (AUCeffect) measured by either method did not reflect the variations in plasma AUC across individuals. The individual variability in AUCeffect from the EEG was similar to that measured by the DSST. CONCLUSIONS Both the EEG and the DSST reflect the central benzodiazepine agonist effects of triazolam. Intrinsic variability in both measures is similar.
Collapse
Affiliation(s)
- David J Greenblatt
- Department of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine and Tufts-New England Medical Center, Boston, MA 02111, USA.
| | | | | | | |
Collapse
|
36
|
Venkatakrishnan K, Culm KE, Ehrenberg BL, Harmatz JS, Corbett KE, Fleishaker JC, Greenblatt DJ. Kinetics and dynamics of intravenous adinazolam, N-desmethyl adinazolam, and alprazolam in healthy volunteers. J Clin Pharmacol 2005; 45:529-37. [PMID: 15831776 DOI: 10.1177/0091270004269105] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The pharmacokinetics and pharmacodynamics of adinazolam mesylate (10 mg), N-desmethyl adinazolam mesylate (NDMAD, 10 mg), and alprazolam (1 mg) were investigated in 9 healthy male subjects in a randomized, blinded, single-dose, 4-way crossover study. All drugs were intravenously infused over 30 minutes. Plasma adinazolam, NDMAD, and alprazolam concentrations, electroencephalographic (EEG) activity in the beta (12-30 Hz) range, performance on the Digit Symbol Substitution Test (DSST), and subjective measures of mood and sedation were monitored for 12 to 24 hours. Mean pharmacokinetic parameters for adinazolam, NDMAD, and alprazolam, respectively, were as follows: volume of distribution (L), 106, 100, and 77; elimination half-life (hours), 2.9, 2.8, and 14.6; and clearance (mL/min), 444, 321, and 84. More than 80% of the total infused adinazolam dose was converted to systemically appearing NDMAD. All 3 benzodiazepine agonists significantly increased beta EEG activity, with alprazolam showing the strongest agonist activity and adinazolam showing the weakest activity. Alprazolam and NDMAD significantly decreased DSST performance, whereas adinazolam had no effect relative to placebo. Adinazolam, NDMAD, and alprazolam all produced significant observer-rated sedation. Plots of EEG effect versus plasma alprazolam concentration demonstrated counterclockwise hysteresis, consistent with an effect site delay. This was incorporated into a kinetic-dynamic model in which hypothetical effect site concentration was related to pharmacodynamic EEG effect via the sigmoid E(max) model, yielding an effect site equilibration half-life of 4.8 minutes. The exponential effect model described NDMAD pharmacokinetics and EEG pharmacodynamics. The relation of both alprazolam and NDMAD plasma concentrations to DSST performance could be described by a modified exponential model. Pharmacokinetic-dynamic modeling was not possible for adinazolam, as the data did not conform to any known concentration-effect model. Collectively, these results indicate that the benzodiazepine-like effects occurring after adinazolam administration are mediated by mainly NDMAD.
Collapse
Affiliation(s)
- Karthik Venkatakrishnan
- Department of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Connemann BJ, Mann K, Lange-Asschenfeldt C, Ruchsow M, Schreckenberger M, Bartenstein P, Gründer G. Anterior limbic alpha-like activity: a low resolution electromagnetic tomography study with lorazepam challenge. Clin Neurophysiol 2005; 116:886-94. [PMID: 15792898 DOI: 10.1016/j.clinph.2004.11.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2004] [Revised: 10/23/2004] [Accepted: 11/15/2004] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To verify findings of an independently regulated anterior limbic alpha band source. METHODS In a randomised cross-over study, the spontaneous EEG was recorded in nine healthy subjects after i.v. lorazepam or placebo. Intracerebral current densities within classical frequency bands were estimated with low resolution electromagnetic tomography [LORETA] and compared between groups with t-statistical parametric mapping [SPM[t]]. A region-of-interest [ROI] based method was used to compare frontal and occipital alpha band activity changes. RESULTS Irrespective of treatment group, local maxima of alpha band power were localised both in the occipital lobe, Brodman area [BA] 18, and in the anterior cingulate cortex [ACC], BA 32. Statistical parametric mapping showed reduced parieto-occipital, but unaltered frontal alpha band power after lorazepam. This result was confirmed by ROI-based comparison of BA 18 and BA 32. CONCLUSIONS There was an anterior limbic maximum of alpha band activity which, unlike occipital alpha, was not suppressed by lorazepam. SIGNIFICANCE The well-known anterior alpha band components may originate from a narrowly circumscribed source, located in the ACC. Frontal and occipital alpha band activities appear to be independently regulated.
Collapse
Affiliation(s)
- Bernhard J Connemann
- Department of Psychiatry III, University of Ulm, Leimgrubenweg 12, 89075 Ulm, Germany.
| | | | | | | | | | | | | |
Collapse
|
38
|
Kabbaj M, Vachon P, Varin F. Impact of peripheral elimination on the concentration-effect relationship of remifentanil in anaesthetized dogs. Br J Anaesth 2005; 94:357-65. [PMID: 15653707 DOI: 10.1093/bja/aei058] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND This study elucidates the impact of sampling site when estimating pharmacokinetic-pharmacodynamic (PK-PD) parameters of drugs such as remifentanil that undergo tissue extraction in the biophase. The interrelationship between the concentrations of remifentanil predicted for the effect compartment and those measured in arterial, venous, and cerebrospinal fluid were investigated under steady-state conditions. METHODS Following induction of anaesthesia with pentobarbital, an arterial cannula (femoral) and two venous catheters (jugular and femoral) were inserted. Electrodes were placed for EEG recording of theta wave activity. Each dog received two consecutive 5-min infusions for the PK-PD study and a bolus followed by a 60-min infusion was started for the steady-state study. Cerebrospinal fluid, arterial and venous blood samples were drawn simultaneously after 30, 40, and 50 min. At the end of the infusion, arterial blood samples were collected for pharmacokinetic analysis. RESULTS Remifentanil PK-PD parameters based on theta wave activity were as follows: apparent volume of distribution at steady-state (V(ss)) (231+/-37 ml kg(-1)), total body clearance (Cl) (63+/-16 ml min(-1) kg(-1)), terminal elimination half-life (t(1/2 beta)) (7.71 min), effect compartment concentration at 50% of maximal observed effect (EC(50)) (21+/-13 ng ml(-1)), and equilibration rate constant between plasma and effect compartment (k(e0)) (0.48+/-0.24 min). The mean steady-state cerebrospinal fluid concentration of 236 ng ml(-1) represented 52 and 74% of that in arterial and venous blood, respectively. CONCLUSIONS Our study re-emphasizes the importance of a sampling site when performing PK-PD modelling for drugs undergoing elimination from the effect compartment. For a drug undergoing tissue elimination such as remifentanil, venous rather than arterial concentrations will reflect more exactly the effect compartment concentrations, under steady-state conditions.
Collapse
Affiliation(s)
- M Kabbaj
- Faculté de Pharmaci, Université de Montréal, Montréal, Québec, Canada
| | | | | |
Collapse
|
39
|
Greenblatt DJ, Ehrenberg BL, Culm KE, Scavone JM, Corbett KE, Friedman HL, Harmatz JS, Shader RI. Kinetics and EEG Effects of Midazolam during and after 1-Minute, 1-Hour, and 3-Hour Intravenous Infusions. J Clin Pharmacol 2004; 44:605-11. [PMID: 15145968 DOI: 10.1177/0091270004265368] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The objective of this study was to evaluate the kinetics and dynamics of midazolam when administered by three different infusion schemes, using electroencephalography to measure pharmacodynamic effects. In a three-way crossover study, 8 volunteers received midazolam (0.1 mg/kg) by constant-rate intravenous infusion. The durations of midazolam infusions for the three trials were 1 minute, 1 hour, and 3 hours. Plasma midazolam concentrations and electroencephalographic (EEG) activity in the 13- to 30-Hz range were monitored for 24 hours. Based on separate analysis of each subject-trial, mean values for volume of distribution and distribution or elimination half-life did not significantly vary. Central compartment volume and clearance differed among the three midazolam infusion trials; however, the magnitude of change was small. EEG activity in the 13- to 30-Hz range significantly increased for all three midazolam infusion trials. Plots of midazolam plasma concentration versus pharmacodynamic EEG effect for the 1-hour and 3-hour infusion trials did not reveal evidence of either counterclockwise or clockwise hysteresis. Plots from the 1-minute infusion trial demonstrated counterclockwise hysteresis, consistent with an equilibration effect-site delay. This was incorporated into a kinetic-dynamic model in which hypothetical effect-site concentration was related to pharmacodynamic EEG effect via the sigmoid E(max) model. Analysis of all three infusion trials together yielded the following mean estimates: maximum EEG effect, 16.3% over baseline; 50% maximum effective concentration, 31 ng/mL; and an apparent rate constant for drug disappearance from the effect compartment which approached infinity. Despite the delay in effect onset during the 1-minute midazolam infusion, midazolam infusions in duration of up to 3 hours produce CNS sedation without evidence of tolerance.
Collapse
Affiliation(s)
- David J Greenblatt
- Department of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
van Ruth S, Mathôt RAA, Sparidans RW, Beijnen JH, Verwaal VJ, Zoetmulder FAN. Population pharmacokinetics and pharmacodynamics of mitomycin during intraoperative hyperthermic intraperitoneal chemotherapy. Clin Pharmacokinet 2004; 43:131-43. [PMID: 14748621 DOI: 10.2165/00003088-200443020-00005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND During recent years, cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy (HIPEC) with mitomycin has been used for various malignancies. OBJECTIVE To characterise the population pharmacokinetics and pharmacodynamics of mitomycin during HIPEC. METHODS Forty-seven patients received mitomycin 35 mg/m2 intraperitoneally as a perfusion over 90 minutes. Mitomycin concentrations were determined in both the peritoneal perfusate and plasma. The observed concentration-time profiles were used to develop a population pharmacokinetic model using nonlinear mixed-effect modelling (NONMEM). The area under the plasma concentration-time curve (AUC) was related to the haematological toxicity. RESULTS Concentration-time profiles of mitomycin in perfusate and plasma were adequately described with one- and two-compartment models, respectively. The average volume of distribution of the perfusate compartment (V1) and rate constant from the perfusate to the systemic circulation (k12) were 4.5 +/- 1.1L and 0.014 +/- 0.003 min(-1), respectively (mean +/- SD, n = 47). The average volume of distribution of the central plasma compartment (V2), clearance from the central compartment (CL) and volume of distribution of the peripheral plasma compartment (V3) were 28 +/- 16L, 0.55 +/- 0.18 L/min and 36 +/- 8L, respectively. The relationship between the AUC in plasma and degree of leucopenia was described with a sigmoidal maximum-effect (Emax) model. CONCLUSIONS The pharmacokinetics of mitomycin during HIPEC could be fitted successfully to a multicompartment model. Relationships between plasma exposure and haematological toxicity were quantified. The developed pharmacokinetic-pharmacodynamic model can be used to simulate different dosage schemes in order to optimise mitomycin administration during HIPEC.
Collapse
Affiliation(s)
- Serge van Ruth
- Department of Surgical Oncology, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
41
|
Jonker DM, Vermeij DAC, Edelbroek PM, Voskuyl RA, Piotrovsky VK, Danhof M. Pharmacodynamic analysis of the interaction between tiagabine and midazolam with an allosteric model that incorporates signal transduction. Epilepsia 2003; 44:329-38. [PMID: 12614388 DOI: 10.1046/j.1528-1157.2003.37802.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PURPOSE The objective of this study was to characterize quantitatively the pharmacodynamic interaction between midazolam (MDL), an allosteric modulator of the gamma-aminobutyric acid subtype A (GABAA) receptor, and tiagabine (TGB), an inhibitor of synaptic GABA uptake. METHODS The in vivo concentration-response relation of TGB was determined through pharmacokinetic/pharmacodynamic (PK/PD) modeling. Rats received a single intravenous dose of 10 mg/kg TGB in the absence and the presence of a steady-state plasma concentration of MDL. The EEG response in the 11.5- to 30-Hz frequency band was used as the pharmacodynamic end point. RESULTS Infusion of MDL resulted in a mean steady-state plasma concentration of 66 +/- 3 ng/ml. A significant pharmacokinetic interaction with TGB was observed. MDL inhibited TGB clearance by 20 +/- 7 ml/min/kg from the original value of 89 +/- 6 ml/min/kg. However, no changes in plasma protein binding of both drugs were observed. The concentration-EEG relation of TGB was described by the sigmoid-Emax model. The pharmacodynamic parameter estimates of TGB were: Emax = 327 +/- 10 microV, EC50 = 392 +/- 20 ng/ml, and nH = 3.1 +/- 0.3. These values were not significantly different in the presence of MDL. Factors that may explain the lack of synergism were identified by a mechanism-based interaction model that separates the receptor activation from the signal-transduction process. High efficiency of signal transduction and the presence of a baseline response were shown to diminish the degree of synergism. CONCLUSIONS We conclude that the in vivo pharmacodynamic interaction between MDL and TGB is additive rather than synergistic. This strongly suggests that allosteric modulation of the antiseizure activity of a GAT-1 inhibitor by a benzodiazepine does not offer a therapeutic advantage.
Collapse
Affiliation(s)
- Daniël M Jonker
- Leiden/Amsterdam Center for Drug Research, Division of Pharmacology, Gorlaeus Laboratory, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
42
|
Visser SAG, Wolters FLC, van der Graaf PH, Peletier LA, Danhof M. Dose-dependent EEG effects of zolpidem provide evidence for GABA(A) receptor subtype selectivity in vivo. J Pharmacol Exp Ther 2003; 304:1251-7. [PMID: 12604703 DOI: 10.1124/jpet.102.044859] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Zolpidem is a nonbenzodiazepine GABA(A) receptor modulator that binds in vitro with high affinity to GABA(A) receptors expressing alpha(1) subunits but with relatively low affinity to receptors expressing alpha(2), alpha(3), and alpha(5) subunits. In the present study, it was investigated whether this subtype selectivity could be detected and quantified in vivo. Three doses (1.25, 5, and 25 mg) of zolpidem were administered to rats in an intravenous infusion over 5 min. The time course of the plasma concentrations was determined in conjunction with the change in the beta-frequency range of the EEG as pharmacodynamic endpoint. The concentration-effect relationship of the three doses showed a dose-dependent maximum effect and a dose-dependent potency. The data were analyzed for one- or two-site binding using two pharmacodynamic models based on 1) the descriptive model and 2) a novel mechanism-based pharmacokinetic/pharmacodynamic (PK/PD) model for GABA(A) receptor modulators that aims to separates drug- and system-specific properties, thereby allowing the estimation of in vivo affinity and efficacy. The application of two-site models significantly improved the fits compared with one-site models. Furthermore, in contrast to the descriptive model, the mechanism-based PK/PD model yielded dose-independent estimates for affinity (97 +/- 40 and 33,100 +/- 14,800 ng x ml(-1)). In conclusion, the mechanism-based PK/PD model is able to describe and explain the observed dose-dependent EEG effects of zolpidem and suggests the subtype selectivity of zolpidem in vivo.
Collapse
Affiliation(s)
- S A G Visser
- Division of Pharmacology, Leiden/Amsterdam Center for Drug Research, Leiden University, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
43
|
Edwards SR, Mather LE. Alfentanil potentiates anaesthetic and electroencephalographic responses to ketamine in the rat. Eur J Pharmacol 2003; 460:27-35. [PMID: 12535856 DOI: 10.1016/s0014-2999(02)02827-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The interactions between mu-opioid and N-methyl-D-aspartate (NMDA) receptors have important implications for clinical pain management. We recently examined the pharmacokinetics of ketamine in rats following i.v. infusion of ketamine (racemate, 50 mg/kg/5 min) and found increased central nervous system distribution of ketamine in the presence of low constant plasma alfentanil concentrations (approximately 50 ng/ml). We now report on the effects of low plasma alfentanil concentrations on the duration of anaesthetic and electroencephalographic (EEG) responses to i.v. infusion of ketamine. Compared to ketamine alone, alfentanil significantly increased both the duration of anaesthesia (by 130%, P=0.00022) and the processed EEG power (microV(2)/s) (by 48%, P=0.0040). The plasma ketamine concentration producing half-maximal EEG effect was significantly reduced (by 60%, P<0.0001) in the presence of alfentanil. The results indicate that low plasma alfentanil concentrations potentiate the anaesthetic and EEG effects produced by ketamine.
Collapse
Affiliation(s)
- Stephen R Edwards
- Centre for Anaesthesia and Pain Management Research, University of Sydney at Royal North Shore Hospital, St. Leonard's NSW, 2065, Australia
| | | |
Collapse
|
44
|
Abstract
Despite the growing means devoted to research and development (R α D) and refinements in the preclinical stages, the efficiency of central nervous system (CMS) drug development is disappointing. Many drugs reach patient studies with an erroneous therapeutic indication andlor in incorrect doses. Apart from the first clinical studies, which are conducted in healthy volunteers and focus only on safety, iolerability, and pharmacokinetics, drug development mostly relies on patient studies. Psychiatric disorders are characterized by heterogeneity and a high rate of comorbidity. It is becoming increasingly difficult to recruit patients for clinical trials and there are many confounding factors in this population, for example, those related to treatments. In order to keep patient exposure and financial expenditure to a minimum, it is important to avoid ill-designed and inconclusive studies. This risk could be minimized by gathering pharmacodynamic data earlier in development and considering that the goal of a phase 1 plan is to reach patient studies with clear ideas about the compound's pharmacodynamic profile, its efficacy in the putative indication (proof of concept), and pharmacokinetic/pharmacodynamic relationships, in addition to safety, tolerability, and pharmacokinetics. Human models in healthy volunteers may be useful tools for this purpose, but their use necessitates a global adaptation of the phase scheme, favoring pharmacodynamic assessments without neglecting safety. We are engaged in an R α D program aimed to adapt existing models and develop new paradigms suitable for early proof of concept substantiation.
Collapse
|
45
|
de Lange ECM, Danhof M. Considerations in the use of cerebrospinal fluid pharmacokinetics to predict brain target concentrations in the clinical setting: implications of the barriers between blood and brain. Clin Pharmacokinet 2002; 41:691-703. [PMID: 12162757 DOI: 10.2165/00003088-200241100-00001] [Citation(s) in RCA: 179] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In the clinical setting, drug concentrations in cerebrospinal fluid (CSF) are sometimes used as a surrogate for drug concentrations at the target site within the brain. However, the brain consists of multiple compartments and many factors are involved in the transport of drugs from plasma into the brain and the distribution within the brain. In particular, active transport processes at the level of the blood-brain barrier and blood-CSF barrier, such as those mediated by P-glycoprotein, may lead to complex relationships between concentrations in plasma, ventricular and lumbar CSF, and other brain compartments. Therefore, CSF concentrations may be difficult to interpret and may have limited value. Pharmacokinetic data obtained by intracerebral microdialysis monitoring may be used instead, providing more valuable information. As non-invasive alternative techniques, positron emission tomography or magnetic resonance spectroscopy may be of added value.
Collapse
Affiliation(s)
- Elizabeth C M de Lange
- Division of Pharmacology, Leiden/Amsterdam Center for Drug Research, Sylvius Laboratories, Leiden, The Netherlands.
| | | |
Collapse
|
46
|
Kotegawa T, Laurijssens BE, Von Moltke LL, Cotreau MM, Perloff MD, Venkatakrishnan K, Warrington JS, Granda BW, Harmatz JS, Greenblatt DJ. In vitro, pharmacokinetic, and pharmacodynamic interactions of ketoconazole and midazolam in the rat. J Pharmacol Exp Ther 2002; 302:1228-37. [PMID: 12183684 DOI: 10.1124/jpet.102.035972] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Interactions of midazolam and ketoconazole were studied in vivo and in vitro in rats. Ketoconazole (total dose of 15 mg/kg intraperitoneally) reduced clearance of intravenous midazolam (5 mg/kg) from 79 to 55 ml/min/kg (p < 0.05) and clearance of intragastric midazolam (15 mg/kg) from 1051 to 237 ml/min/kg (p < 0.05), increasing absolute bioavailability from 0.11 to 0.36 (p < 0.05). Presystemic extraction occurred mainly across the liver as opposed to the gastrointestinal tract mucosa. Midazolam increased electroencephalographic (EEG) amplitude in the beta-frequency range. Ketoconazole shifted the concentration-EEG effect relationship rightward (increase in EC(50)), probably because ketoconazole is a neutral benzodiazepine receptor ligand. Ketoconazole competitively inhibited midazolam hydroxylation by rat liver and intestinal microsomes in vitro, with nanomolar K(i) values. At a total serum ketoconazole of 2 microg/ml (3.76 microM) in vivo, the predicted reduction in clearance of intragastric midazolam by ketoconazole (to 6% of control) was slightly greater than the observed reduction in vivo (to 15% of control). However, unbound serum ketoconazole greatly underpredicted the observed clearance reduction. Although the in vitro and in vivo characteristics of midazolam in rats incompletely parallel those in humans, the experimental model can be used to assess aspects of drug interactions having potential clinical importance.
Collapse
Affiliation(s)
- Tsutomu Kotegawa
- Department of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, Tufts-New England Medical Center, 136 Harrison Avenue, Boston, MA 02111, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wang L, Wang XD. Pharmacokinetic and pharmacodynamic effects of clonazepam in children with epilepsy treated with valproate: a preliminary study. Ther Drug Monit 2002; 24:532-6. [PMID: 12142639 DOI: 10.1097/00007691-200208000-00012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The authors report the use of the quantitative pharmaco-EEG (QPEEG) technique to study the pharmacokinetics (PK) and pharmacodynamics (PD) of clonazepam (CZP) in four epileptic children who suffered uncontrolled seizures despite long-term valproate (VPA) therapy. After a single dose of CZP (0.05 mg/kg, PO), blood samples were collected at 0, 0.5, 1.0, 1.5, 2.0, 3.0, 4.0, 8.0, 12.0, and 24.0 hours. CZP and VPA concentrations were measured by HPLC or GC assay, respectively. At each blood collection time point, EEG signals (60 s) were recorded for brain electrical activity mapping, and the power percentage average (PPA) of each frequency band was calculated. The relationship between drug concentrations and their corresponding PPA of each frequency band was analyzed. VPA steady-state drug concentrations (Css) were within the therapeutic range and not affected by CZP. The peak concentration (Cmax) of CZP and the time intervals from dosing to Cmax (Tmax) were 20.9 ng/mL to 113.8 ng/mL and 1 hour to 1.5 hours, respectively. There was no significant correlation between VPA concentrations and the PPA of any of the EEG frequency bands. CZP blood concentrations showed significant correlation with PPA in 3 of the 4 patients. Our results suggested CZP could affect fast wave activities in proportion to CZP blood concentrations. We propose that QPEEG is a promising technique to study the PK and PD of selected anti-epileptic drugs.
Collapse
Affiliation(s)
- Li Wang
- Pediatric Neurology, Peking University First Hospital, Beijing, China.
| | | |
Collapse
|
48
|
Tuk B, van Gool T, Danhof M. Mechanism-based pharmacodynamic modeling of the interaction of midazolam, bretazenil, and zolpidem with ethanol. J Pharmacokinet Pharmacodyn 2002; 29:235-50. [PMID: 12449497 DOI: 10.1023/a:1020202806759] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The pharmacokinetic and pharmacodynamic interactions of ethanol with the full benzodiazepine agonist midazolam, the partial agonist bretazenil and the benzodiazepine BZ1 receptor subtype selective agonist zolpidem have been determined in the rat in vivo, using an integrated pharmacokinetic-pharmacodynamic approach. Ethanol was administered as a constant rate infusion resulting in constant plasma concentrations of 0.5 g/l. The pharmacokinetics and pharmacodynamics of midazolam, bretazenil, and zolpidem were determined following an intravenous infusion of 5.0, 2.5, and 18 mg/kg respectively. The amplitude in the 11.5-30 Hz frequency band of the EEG was used as measure of the pharmacological effect. For each of the benzodiazepines the concentration-EEG effect relationship could be described by the sigmoid Emax pharmacodynamic model. Significant differences in both EC50 and Emax were observed. The values of the EC50 were 76 +/- 11, 12 +/- 3, and 512 +/- 116 ng/ml for midazolam, bretazenil, and zolpidem respectively. The values of the Emax were 113 +/- 9, 44 +/- 3, and 175 +/- 10 microV/s. In the presence of ethanol the values of the EC50 of midazolam and zolpidem were reduced to approximately 50% of the original value. The values for Emax and Hill-factor were unchanged Due to a large interindividual variability no significant change in EC50 was observed for bretazenil. Analysis of the data on basis of a mechanism-based model showed only a decrease in the apparent affinity constant KPD for all three drugs, indicating that changes in EC50 can be explained entirely by a change in the apparent affinity constant KPD without concomitant changes in the efficacy parameter ePD and the stimulus-effect relationship. The findings of this study show that the pharmacodynamic interactions with a low dose of ethanol in vivo are qualitatively and quantitatively similar for benzodiazepine receptor full agonists, partial agonists, and benzodiazepine BZ1 receptor subtype selective agonists. This interaction can be explained entirely by a change in the affinity of the biological system for each benzodiazepine.
Collapse
Affiliation(s)
- Bert Tuk
- Leiden/Amsterdam Center for Drug Research, Division of Pharmacology, Leiden University, PO Box 9502, 2300 RA Leiden, The Netherlands
| | | | | |
Collapse
|
49
|
Ohtani Y, Kotegawa T, Tsutsumi K, Morimoto T, Hirose Y, Nakano S. Effect of fluconazole on the pharmacokinetics and pharmacodynamics of oral and rectal bromazepam: an application of electroencephalography as the pharmacodynamic method. J Clin Pharmacol 2002; 42:183-91. [PMID: 11831541 DOI: 10.1177/00912700222011229] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Quantitative analysis of electroencephalography (EEG) is used increasingly to evaluate the pharmacodynamics of benzodiazepines. The present study aimed to apply the EEG method as well as more traditional approaches to an interaction study of bromazepam and fluconazole. Twelve healthy male volunteers participated in a randomized, double-blind, four-way crossover study. The subjects received single oral or rectal doses of bromazepam (3 mg) after 4-day pretreatment of oral fluconazole (100 mg daily) or its placebo. Plasma bromazepam concentrations were measured before and 0.5, 1, 2, 3, 4, 6, 12, 22, 46, and 70 hours after bromazepam administration. Pharmacodynamic effects of bromazepam were assessed using self-rated drowsiness, continuous number addition test, and EEG. Fluconazole caused no significant changes in pharmacokinetics and pharmacodynamics of oral or rectal bromazepam. Rectal administration significantly increased AUC (1.7-fold, p < 0.0001) and Cmax (1.6-fold, p < 0.0001) of bromazepam. These changes following rectal dose may be due to avoidance of degradation occurring in the gastrointestinal tract. Rectal bromazepam also increased the area under the effect curves assessed by EEG (p < 0.05) and subjective drowsiness (p < 0.05). EEG effects were closely correlated with mean plasma bromazepam concentrations (r = 0.92, p < 0.001 for placebo; r = 0.89, p < 0.0001 for fluconazole). Thus, the EEG method provided pharmacodynamic data that clearly reflected the pharmacokinetics of bromazepam.
Collapse
Affiliation(s)
- Yasukiyo Ohtani
- Department of Clinical Pharmacology and Therapeutics, Oita Medical University, Japan
| | | | | | | | | | | |
Collapse
|
50
|
Laurijssens BE, Greenblatt DJ. Effect of 7-day exposure to midazolam on electroencephalogram pharmacodynamics in rats: a model to study multiple pharmacokinetic-pharmacodynamic relationships in individual animals. J Pharm Pharmacol 2002; 54:77-86. [PMID: 11829133 DOI: 10.1211/0022357021771959] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The objective of this study was to determine the concentration-electroencephalogram (EEG) relationships for midazolam, a full-agonist benzodiazepine ligand, on multiple occasions in individual rats, and to examine the effect of chronic midazolam exposure on that relationship. Rats were chronically instrumented with venous and arterial cannulas, and cortical EEG electrodes. The rats received either: 7 days of midazolam 10 mg kg(-1) intravenously once a day (midazolam group); or midazolam on days 1 and 7 and vehicle on days 2-6 (vehicle group). Concentration-effect relationships were determined on days 1, 4 and 7 from multiple blood and EEG samples before and after the administration of the midazolam dose. The concentration-EEG effect relationships were consistent with a sigmoidal Emax (maximal effect) model. No differences in pharmacokinetic or pharmacodynamic parameters were found between day 1 and day 7 in either group. However, in the midazolam group, both the fraction unbound of midazolam in serum and the EC50 (concentration at half-maximal effect) for free midazolam increased from days 1-7 by 35 +/- 3% and 54 +/- 25%, respectively (means +/- s.d., P< 0.05). This may be related to decreased serum albumin levels in the midazolam group (-19+5%, P < 0.05) which, in turn, could be explained by the sedation associated with daily midazolam treatment. We concluded that concentration-EEG effect relationships can be studied on multiple occasions in individual animals, reducing animal use and variability. A modest degree of tolerance to midazolam was found with this paradigm, the effect only being evident after correction for the fraction unbound of midazolam.
Collapse
Affiliation(s)
- Bart E Laurijssens
- Department of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, Boston, MA 02111, USA
| | | |
Collapse
|