Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
J Pharmacol Exp Ther ; 383(3): 238-245, 2022 12.
Article in English | MEDLINE | ID: mdl-36167415

ABSTRACT

Although propofol is among the most commonly administered general anesthetics, its mechanism of action is not fully understood. It has been hypothesized that propofol acts via a similar mechanism as (R)-ethyl 1-(1-phenylethyl)-1H-imidazole-5-carboxylate (etomidate) by binding within the GABAA receptor transmembrane receptor domain at the two ß +/α - subunit interfaces with resultant positive allosteric modulation. To test this hypothesis, we leveraged the ability of diazepam to bind to those sites and act as a competitive antagonist. We used oocyte-expressed α 1 ß 3 γ 2L GABAA receptors to define the actions of diazepam (± flumazenil) on currents activated or potentiated by propofol and a zebrafish activity assay to define the impact of diazepam and flumazenil on propofol-induced anesthesia. We found that diazepam increased the amplitudes of GABAA receptor-mediated currents at nanomolar concentrations but reduced them at micromolar concentrations. The current amplitude changes produced by nanomolar diazepam concentrations were inhibited by flumazenil whereas those produced by micromolar diazepam concentrations were not. Studies of agonist potentiation showed that the micromolar inhibitory action of diazepam was surmountable by high concentrations of propofol and produced a rightward shift in the propofol concentration-response curve characterized by a Schild slope not statistically significantly different from 1, consistent with competition between diazepam and propofol. Although micromolar concentrations of diazepam (plus flumazenil) similarly reduced GABAA receptor currents modulated by propofol and etomidate, it only reduced the anesthetic actions of etomidate. We conclude that while both propofol and etomidate can modulate GABAA receptors by binding to the ß +/α - subunit interfacial sites, propofol-induced anesthesia likely involves additional target sites. SIGNIFICANCE STATEMENT: Although the drug combination of diazepam and flumazenil reverses the GABAA receptor positive modulatory actions of both propofol and (R)-ethyl 1-(1-phenylethyl)-1H-imidazole-5-carboxylate (etomidate), it only reverses the in vivo anesthetic actions of etomidate. These results strongly suggest that distinct mechanisms of action account for the anesthetic actions of these two commonly administered anesthetic agents.


Subject(s)
Etomidate , Propofol , Animals , Receptors, GABA-A/metabolism , Propofol/pharmacology , Diazepam/pharmacology , Zebrafish/metabolism , Etomidate/pharmacology , gamma-Aminobutyric Acid/pharmacology
2.
Ther Adv Endocrinol Metab ; 13: 20420188211058583, 2022.
Article in English | MEDLINE | ID: mdl-35186251

ABSTRACT

Cushing's syndrome (CS) is an endocrine disease characterized by excessive adrenocortical steroid production. One of the mainstay pharmacological treatments for CS are steroidogenesis enzyme inhibitors, including the antifungal agent ketoconazole along with metyrapone, mitotane, and aminoglutethimide. Recently, osilodrostat was added to this drug class and approved by the US Food and Drug Administration (FDA) for the treatment of Cushing's Disease. Steroidogenesis enzyme inhibitors inhibit various enzymes along the cortisol biosynthetic pathway and may be used preoperatively to lower cortisol levels and reduce surgical risk associated with tumor resection or postoperatively when surgery and/or radiation therapies are not curative. Because their selectivities for steroidogenic enzymes vary, they may even be administered in combination to achieve relatively rapid control of severe hypercortisolemia. Unfortunately, all currently available inhibitors are accompanied by serious adverse side effects that limit dosing and often result in treatment failures. Although more commonly known as a general anesthetic induction agent, etomidate is another member of the steroidogenesis enzyme inhibitor drug class. It suppresses cortisol production primarily by inhibiting 11ß-hydroxylase and is the only inhibitor that may be given parenterally. However, the sedative-hypnotic actions of etomidate limit its use as an acute management option for CS. Thus, some have recommended that it be used only in intensive care settings. In this review, we discuss the initial development of etomidate as an anesthetic agent, its subsequent development as a treatment for CS, and the recent advances in dosing and drug development that dissociate sedative-hypnotic and adrenostatic drug actions to facilitate CS treatment in non-critical care settings.

3.
Br J Pharmacol ; 178(24): 4842-4858, 2021 12.
Article in English | MEDLINE | ID: mdl-34386973

ABSTRACT

BACKGROUND AND PURPOSE: In addition to binding to the classical high-affinity extracellular benzodiazepine binding site of the GABAA receptor, some benzodiazepines occupy transmembrane inter-subunit anaesthetic sites that bind etomidate (ß+ /α- sites) or the barbiturate derivative R-mTFD-MPAB (α+ /ß- and γ+ /ß- sites). We aimed to define the functional effects of these interactions on GABAA receptor activity and animal behaviour. EXPERIMENTAL APPROACH: With flumazenil blocking classical high-affinity extracellular benzodiazepine site effects, modulation of GABA-activated currents by diazepam, midazolam and flurazepam was measured electrophysiologically in wildtype and M2-15' mutant α1 ß3 γ2L GABAA receptors. Zebrafish locomotive activity was also assessed in the presence of each benzodiazepine plus flumazenil. KEY RESULTS: In the presence of flumazenil, micromolar concentrations of diazepam and midazolam both potentiated and inhibited wildtype GABAA receptor currents. ß3 N265M (M2-15' in the ß+ /α- sites) and α1 S270I (M2-15' in the α+ /ß- site) mutations reduced or abolished potentiation by these drugs. In contrast, the γ2 S280W mutation (M2-15' in the γ+ /ß- site) abolished inhibition. Flurazepam plus flumazenil only inhibited wildtype receptor currents, an effect unaltered by M2-15' mutations. In the presence of flumazenil, zebrafish locomotion was enhanced by diazepam at concentrations up to 30 µM and suppressed at 100 µM, suppressed by midazolam and enhanced by flurazepam. CONCLUSIONS AND IMPLICATIONS: Benzodiazepine binding to transmembrane anaesthetic binding sites of the GABAA receptor can produce positive or negative modulation manifesting as decreases or increases in locomotion, respectively. Selectivity for these sites may contribute to the distinct GABAA receptor and behavioural actions of different benzodiazepines, particularly at high (i.e. anaesthetic) concentrations.


Subject(s)
Anesthetics , Receptors, GABA-A , Animals , Anesthetics/pharmacology , Benzodiazepines/metabolism , Benzodiazepines/pharmacology , Binding Sites , Flumazenil/chemistry , Flumazenil/pharmacology , Receptors, GABA-A/metabolism , Zebrafish/metabolism
5.
Anesthesiology ; 133(3): 583-594, 2020 09.
Article in English | MEDLINE | ID: mdl-32541553

ABSTRACT

BACKGROUND: Recent cryo-electron microscopic imaging studies have shown that in addition to binding to the classical extracellular benzodiazepine binding site of the α1ß3γ2L γ-aminobutyric acid type A (GABAA) receptor, diazepam also binds to etomidate binding sites located in the transmembrane receptor domain. Because such binding is characterized by low modulatory efficacy, the authors hypothesized that diazepam would act in vitro and in vivo as a competitive etomidate antagonist. METHODS: The concentration-dependent actions of diazepam on 20 µM etomidate-activated and 6 µM GABA-activated currents were defined (in the absence and presence of flumazenil) in oocyte-expressed α1ß3γ2L GABAA receptors using voltage clamp electrophysiology. The ability of diazepam to inhibit receptor labeling of purified α1ß3γ2L GABAA receptors by [H]azietomidate was assessed in photoaffinity labeling protection studies. The impact of diazepam (in the absence and presence of flumazenil) on the anesthetic potencies of etomidate and ketamine was compared in a zebrafish model. RESULTS: At nanomolar concentrations, diazepam comparably potentiated etomidate-activated and GABA-activated GABAA receptor peak current amplitudes in a flumazenil-reversible manner. The half-maximal potentiating concentrations were 39 nM (95% CI, 27 to 55 nM) and 26 nM (95% CI, 16 to 41 nM), respectively. However, at micromolar concentrations, diazepam reduced etomidate-activated, but not GABA-activated, GABAA receptor peak current amplitudes in a concentration-dependent manner with a half-maximal inhibitory concentration of 9.6 µM (95% CI, 7.6 to 12 µM). Diazepam (12.5 to 50 µM) also right-shifted the etomidate-concentration response curve for direct activation without reducing the maximal response and inhibited receptor photoaffinity labeling by [H]azietomidate. When administered with flumazenil, 50 µM diazepam shifted the etomidate (but not the ketamine) concentration-response curve for anesthesia rightward, increasing the etomidate EC50 by 18-fold. CONCLUSIONS: At micromolar concentrations and in the presence of flumazenil to inhibit allosteric modulation via the classical benzodiazepine binding site of the GABAA receptor, diazepam acts as an in vitro and in vivo competitive etomidate antagonist.


Subject(s)
Diazepam/pharmacology , Etomidate/antagonists & inhibitors , Hypnotics and Sedatives/pharmacology , Receptors, GABA/drug effects , Animals , Drug Antagonism , Hypnotics and Sedatives/antagonists & inhibitors , Models, Animal , Zebrafish
6.
Pharmacol Rep ; 72(1): 260-266, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32016849

ABSTRACT

BACKGROUND: Many sedative-hypnotic agents are thought to act by positively modulating γ-aminobutyric acid type A (GABAA) receptors. However, for many agents, the location(s) of the binding site(s) responsible for such receptor modulation is uncertain. We previously developed a low efficacy ligand (naphthalene-etomidate) that binds within a homologous set of hydrophobic cavities located at GABAA receptor subunit interfaces in the transmembrane domain, and thus acts as a competitive antagonist for higher efficacy sedative-hypnotics that also bind to these sites. In this report, we describe studies using this compound as a pharmacological screening tool to test whether sedative-hypnotics representing a range of chemical classes can modulate GABAA receptors by binding within these receptor cavities. METHODS: The impact of naphthalene-etomidate on GABA-evoked currents that were mediated by oocyte-expressed α1ß3γ2L GABAA receptors and potentiated by muscimol, alphaxalone, 2,2,2-trichloroethanol, isoflurane, AA29504, loreclezole, or diazepam was quantified using electrophysiological techniques. RESULTS: Naphthalene-etomidate (300 µM) significantly reduced GABAA receptor currents potentiated by alphaxalone (by 22 ± 11%), 2,2,2-trichloroethanol (by 23 ± 6%), isoflurane (by 32 ± 10%), AA29504 (by 41 ± 6%), loreclezole (by 43 ± 9%), but significantly increased those potentiated by muscimol (by 26 ± 11%). Naphthalene-etomidate significantly increased currents potentiated by a low (1 µM) diazepam concentration (by 56 ± 14%) while reducing those potentiated by a high (100 µM) diazepam concentration (by 11 ± 7%). CONCLUSIONS: Our results suggest that many (but not all) sedative-hypnotics are capable of positively modulating the GABAA receptor by binding within a common set of hydrophobic cavities.


Subject(s)
Etomidate/pharmacology , GABA-A Receptor Antagonists/pharmacology , Hypnotics and Sedatives/administration & dosage , Naphthalenes/pharmacology , Animals , Etomidate/administration & dosage , Female , GABA-A Receptor Antagonists/administration & dosage , Humans , Hydrophobic and Hydrophilic Interactions , Hypnotics and Sedatives/pharmacology , Ligands , Naphthalenes/administration & dosage , Receptors, GABA-A , Xenopus laevis , gamma-Aminobutyric Acid/metabolism
7.
Trends Pharmacol Sci ; 40(11): 809-811, 2019 11.
Article in English | MEDLINE | ID: mdl-31623940

ABSTRACT

All currently available general anesthetics produce potentially deadly side effects. Unfortunately, few approaches have been developed to design safer ones, despite important advances in anesthetic mechanisms research. Cayla and colleagues recently showed that computational methods can be used to identify anesthetic lead compounds devoid of specific side effects.


Subject(s)
Anesthetics , Etomidate , Drug Discovery
8.
BMC Pharmacol Toxicol ; 20(1): 48, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31383012

ABSTRACT

BACKGROUND: Cushing's syndrome is an endocrine disorder characterized by the overproduction of adrenocortical steroids. Steroidogenesis enzyme inhibitors are the mainstays of pharmacological treatment. Unfortunately, they produce significant side effects. Among the most potent inhibitors is the general anesthetic etomidate whose GABAA receptor-mediated sedative-hypnotic actions restrict use. In this study, we defined the sedative-hypnotic and steroidogenesis inhibiting actions of etomidate and four phenyl-ring substituted etomidate analogs (dimethoxy-etomidate, isopropoxy-etomidate, naphthalene-etomidate, and naphthalene (2)-etomidate) that possess negligible GABAA receptor modulatory activities. METHODS: In the first set of experiments, male Sprague-Dawley rats were assessed for loss of righting reflexes (LoRR) after receiving intravenous boluses of either etomidate (1 mg/kg) or an etomidate analog (40 mg/kg). In the second set of experiments, rats were assessed for LoRR and their abilities to produce adrenocortical and androgenic steroids after receiving 2-h infusions (0.5 mg kg- 1 min- 1) of either etomidate or an etomidate analog. RESULTS: All rats that received etomidate boluses or infusions had LoRR that persisted for minutes or hours, respectively. In contrast, no rat that received an etomidate analog had LoRR. Compared to rats in the vehicle control group, rats that received etomidate analog infusions had plasma corticosterone and aldosterone concentrations that were reduced by 80-84% and 68-94%, respectively. Rats that received etomidate infusions had plasma corticosterone and aldosterone concentrations that were also significantly reduced (by 92 and 96%, respectively). Rats that received etomidate or isopropoxy-etomidate had significant reductions (90 and 57%, respectively) in plasma testosterone concentrations whereas those that received naphthalene-etomidate had significant increases (1400%) in plasma dehydroepiandrosterone concentrations. Neither etomidate nor any etomidate analog significantly affected plasma androstenedione and dihydrotestosterone concentrations. CONCLUSIONS: Our studies demonstrate that the four phenyl-ring substituted etomidate analogs form a novel class of compounds that are devoid of sedative-hypnotic activities and suppress plasma concentrations of adrenocortical steroids but vary in their effects on plasma concentrations of androgenic steroids.


Subject(s)
Etomidate/analogs & derivatives , Hypnotics and Sedatives/pharmacology , Steroid Synthesis Inhibitors/pharmacology , Animals , Etomidate/chemistry , Etomidate/pharmacology , Hypnotics and Sedatives/chemistry , Male , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/drug effects , Reflex/drug effects , Steroid Synthesis Inhibitors/chemistry , Steroids/blood
9.
Anesthesiology ; 131(2): 287-304, 2019 08.
Article in English | MEDLINE | ID: mdl-31166238

ABSTRACT

BACKGROUND: The etomidate analog ABP-700 produces involuntary muscle movements that could be manifestations of seizures. To define the relationship (if any) between involuntary muscle movements and seizures, electroencephalographic studies were performed in Beagle dogs receiving supra-therapeutic (~10× clinical) ABP-700 doses. γ-aminobutyric acid type A (GABAA) and glycine receptor studies were undertaken to test receptor inhibition as the potential mechanism for ABP-700 seizures. METHODS: ABP-700 was administered to 14 dogs (6 mg/kg bolus followed by a 2-h infusion at 1 mg · kg(-1) · min(-1), 1.5 mg · kg(-1) · min(-1), or 2.3 mg · kg(-1) · min(-1)). Involuntary muscle movements were documented, electroencephalograph was recorded, and plasma ABP-700 and CPM-acid concentrations were measured during and after ABP-700 administration. The concentration-dependent modulatory actions of ABP-700 and CPM-acid were defined in oocyte-expressed α1ß3γ2L GABAA and α1ß glycine receptors (n = 5 oocytes/concentration) using electrophysiologic techniques. RESULTS: ABP-700 produced both involuntary muscle movements (14 of 14 dogs) and seizures (5 of 14 dogs). However, these phenomena were temporally and electroencephalographically distinct. Mean peak plasma concentrations were (from lowest to highest dosed groups) 35 µM, 45 µM, and 102 µM (ABP-700) and 282 µM, 478 µM, and 1,110 µM (CPM-acid). ABP-700 and CPM-acid concentration-GABAA receptor response curves defined using 6 µM γ-aminobutyric acid exhibited potentiation at low and/or intermediate concentrations and inhibition at high ones. The half-maximal inhibitory concentrations of ABP-700 and CPM-acid defined using 1 mM γ-aminobutyric acid were 770 µM (95% CI, 590 to 1,010 µM) and 1,450 µM (95% CI, 1,340 to 1,560 µM), respectively. CPM-acid similarly inhibited glycine receptors activated by 1 mM glycine with a half-maximal inhibitory concentration of 1,290 µM (95% CI, 1,240 to 1,330 µM). CONCLUSIONS: High dose ABP-700 infusions produce involuntary muscle movements and seizures in Beagle dogs via distinct mechanisms. CPM-acid inhibits both GABAA and glycine receptors at the high (~100× clinical) plasma concentrations achieved during the dog studies, providing a plausible mechanism for the seizures.


Subject(s)
Etomidate/analogs & derivatives , Etomidate/pharmacology , Hypnotics and Sedatives/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , Seizures/chemically induced , Animals , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Electroencephalography/methods , Female , Male
10.
Anesthesiology ; 129(5): 959-969, 2018 11.
Article in English | MEDLINE | ID: mdl-30052529

ABSTRACT

WHAT WE ALREADY KNOW ABOUT THIS TOPIC: WHAT THIS ARTICLE TELLS US THAT IS NEW: BACKGROUND:: Naphthalene-etomidate, an etomidate analog containing a bulky phenyl ring substituent group, possesses very low γ-aminobutyric acid type A (GABAA) receptor efficacy and acts as an anesthetic-selective competitive antagonist. Using etomidate analogs containing phenyl ring substituents groups that range in volume, we tested the hypothesis that this unusual pharmacology is caused by steric hindrance that reduces binding to the receptor's open state. METHODS: The positive modulatory potencies and efficacies of etomidate and phenyl ring-substituted etomidate analogs were electrophysiology defined in oocyte-expressed α1ß3γ2L GABAA receptors. Their binding affinities to the GABAA receptor's two classes of transmembrane anesthetic binding sites were assessed from their abilities to inhibit receptor labeling by the site-selective photolabels [H]azi-etomidate and tritiated R-5-allyl-1-methyl-5-(m-trifluoromethyl-diazirynylphenyl) barbituric acid. RESULTS: The positive modulatory activities of etomidate and phenyl ring-substituted etomidate analogs progressively decreased with substituent group volume, reflecting significant decreases in both potency (P = 0.005) and efficacy (P < 0.0001). Affinity for the GABAA receptor's two ß - α anesthetic binding sites similarly decreased with substituent group volume (P = 0.003), whereas affinity for the receptor's α - ß/γ - ß sites did not (P = 0.804). Introduction of the N265M mutation, which is located at the ß - α binding sites and renders GABAA receptors etomidate-insensitive, completely abolished positive modulation by naphthalene-etomidate. CONCLUSIONS: Steric hindrance selectively reduces phenyl ring-substituted etomidate analog binding affinity to the two ß - α anesthetic binding sites on the GABAA receptor's open state, suggesting that the binding pocket where etomidate's phenyl ring lies becomes smaller as the receptor isomerizes from closed to open.


Subject(s)
Anesthetics, Intravenous/pharmacology , Etomidate/pharmacology , Receptors, GABA/metabolism , Animals , Cell Culture Techniques , Humans , Oocytes , Receptors, GABA/drug effects , Xenopus
11.
Methods Enzymol ; 603: 153-169, 2018.
Article in English | MEDLINE | ID: mdl-29673523

ABSTRACT

All currently available general anesthetic agents possess potentially lethal side effects requiring their administration by highly trained clinicians. Among these agents is etomidate, a highly potent imidazole-based intravenous sedative-hypnotic that deleteriously suppresses the synthesis of adrenocortical steroids in a manner that is both potent and persistent. We developed two distinct strategies to design etomidate analogs that retain etomidate's potent hypnotic activity, but produce less adrenocortical suppression than etomidate. One strategy seeks to reduce binding to 11ß-hydroxylase, a critical enzyme in the steroid biosynthetic pathway, which is potently inhibited by etomidate. The other strategy seeks to reduce the duration of adrenocortical suppression after etomidate administration by modifying the drug's structure to render it susceptible to rapid metabolism by esterases. In this chapter, we describe the methods used to evaluate the hypnotic and adrenocortical inhibitory potencies of two lead compounds designed using the aforementioned strategies. Our purpose is to provide a case study for the development of novel analogs of existing drugs with reduced side effects.


Subject(s)
Anesthetics, Intravenous/pharmacology , Etomidate/pharmacology , Propanolamines/pharmacology , Receptors, GABA-A/chemistry , Remifentanil/pharmacology , Steroid 11-beta-Hydroxylase/chemistry , Adrenal Cortex/drug effects , Adrenal Cortex/metabolism , Adrenocorticotropic Hormone/pharmacology , Anesthetics, Intravenous/chemical synthesis , Animals , Binding Sites , Biotransformation , Drug Discovery , Etomidate/analogs & derivatives , Etomidate/chemical synthesis , Hypnosis, Anesthetic/methods , Hypnotics and Sedatives/chemical synthesis , Hypnotics and Sedatives/pharmacology , Larva/drug effects , Larva/physiology , Molecular Docking Simulation , Propanolamines/chemical synthesis , Protein Binding , Rana pipiens , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Reflex, Righting/drug effects , Reflex, Righting/physiology , Remifentanil/chemical synthesis , Steroid 11-beta-Hydroxylase/metabolism , Structure-Activity Relationship
12.
Anesth Analg ; 126(4): 1241-1248, 2018 04.
Article in English | MEDLINE | ID: mdl-29256939

ABSTRACT

BACKGROUND: Increasing attention has been focused on health care expenditures, which include anesthetic-related drug costs. Using data from 2 large academic medical centers, we sought to identify significant contributors to anesthetic drug cost variation. METHODS: Using anesthesia information management systems, we calculated volatile and intravenous drug costs for 8 types of inpatient surgical procedures performed from July 1, 2009, to December 31, 2011. For each case, we determined patient age, American Society of Anesthesiologists (ASA) physical status, gender, institution, case duration, in-room provider, and attending anesthesiologist. These variables were then entered into 2 fixed-effects linear regression models, both with logarithmically transformed case cost as the outcome variable. The first model included duration, attending anesthesiologist, patient age, ASA physical status, and patient gender as independent variables. The second model included case type, institution, patient age, ASA physical status, and patient gender as independent variables. When all variables were entered into 1 model, redundancy analyses showed that case type was highly correlated (R = 0.92) with the other variables in the model. More specifically, a model that included case type was no better at predicting cost than a model without the variable, as long as that model contained the combination of attending anesthesiologist and case duration. Therefore, because we were interested in determining the effect both variables had on cost, 2 models were created instead of 1. The average change in cost resulting from each variable compared to the average cost of the reference category was calculated by first exponentiating the ß coefficient and subtracting 1 to get the percent difference in cost. We then multiplied that value by the mean cost of the associated reference group. RESULTS: A total of 5504 records were identified, of which 4856 were analyzed. The median anesthetic drug cost was $38.45 (25th percentile = $23.23, 75th percentile = $63.82). The majority of the variation was not described by our models-35.2% was explained in the model containing case duration, and 32.3% was explained in the model containing case type. However, the largest sources of variation our models identified were attending anesthesiologist, case type, and procedure duration. With all else held constant, the average change in cost between attending anesthesiologists ranged from a cost decrease of $41.25 to a cost increase of $95.67 (10th percentile = -$19.96, 90th percentile = +$20.20) when compared to the provider with the median value for mean cost per case. The average change in cost between institutions was significant but minor ($5.73). CONCLUSIONS: The majority of the variation was not described by the models, possibly indicating high per-case random variation. The largest sources of variation identified by our models included attending anesthesiologist, procedure type, and case duration. The difference in cost between institutions was statistically significant but was minor. While many prior studies have found significant savings resulting from cost-reducing interventions, our findings suggest that because the overall cost of anesthetic drugs was small, the savings resulting from interventions focused on the clinical practice of attending anesthesiologists may be negligible, especially in institutions where access to more expensive drugs is already limited. Thus, cost-saving efforts may be better focused elsewhere.


Subject(s)
Anesthetics, Inhalation/economics , Anesthetics, Intravenous/economics , Drug Costs , Health Expenditures , Hospital Costs , Academic Medical Centers/economics , Adult , Aged , Anesthesiologists/economics , Boston , Female , Humans , Male , Middle Aged , Models, Economic , Personnel Staffing and Scheduling/economics , Salaries and Fringe Benefits , Tennessee , Time Factors , Young Adult
13.
J Pharmacol Exp Ther ; 364(2): 229-237, 2018 02.
Article in English | MEDLINE | ID: mdl-29203576

ABSTRACT

Cushing's syndrome is characterized by the overproduction of adrenocortical steroids. Steroidogenesis inhibitors are mainstays of medical therapy for Cushing's syndrome; unfortunately, adverse side effects and treatment failures are common with currently available drugs. The general anesthetic induction agent etomidate is among the most potent inhibitors of adrenocortical steroidogenesis. However, its use as a treatment of Cushing's syndrome is complicated by its sedative-hypnotic activity and ability to produce myoclonus, central nervous system actions thought to be mediated by the GABAA receptor. Here, we describe the pharmacology of the novel etomidate analog (R)-ethyl 1-(1-(3,5-dimethoxyphenyl)ethyl)-1H-imidazole-5-carboxylate (dimethoxy-etomidate). In contrast to etomidate, dimethoxy-etomidate minimally enhanced GABA-evoked GABAA receptor-mediated currents even at a near-saturating aqueous concentration. In Sprague-Dawley rats, dimethoxy-etomidate's potency for producing loss of righting reflexes-an animal model of sedation/hypnosis-was 2 orders of magnitude lower than that of etomidate, and it did not produce myoclonus. However, similar to etomidate, dimethoxy-etomidate potently suppressed adrenocortical steroid synthesis primarily by inhibiting 11ß-hydroxylase. [3H]etomidate binding to rat adrenocortical membranes was inhibited by dimethoxy-etomidate in a biphasic manner with IC50 values of 8.2 and 3970 nM, whereas that by etomidate was monophasic with an IC50 of 22 nM. Our results demonstrate that, similar to etomidate, dimethoxy-etomidate potently and dose-dependently suppresses adrenocortical steroid synthesis by inhibiting 11ß-hydroxylase. However, it is essentially devoid of etomidate's GABAA receptor positive modulatory and sedative-hypnotic activities and produces no myoclonus, providing proof of concept for the design of etomidate analogs without important central nervous system actions for the pharmacologic treatment of Cushing's syndrome.


Subject(s)
Etomidate/analogs & derivatives , Etomidate/pharmacology , Steroids/biosynthesis , Animals , Electrophysiological Phenomena/drug effects , Etomidate/chemistry , Male , Movement/drug effects , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , gamma-Aminobutyric Acid/pharmacology
14.
Anesthesiology ; 127(5): 824-837, 2017 11.
Article in English | MEDLINE | ID: mdl-28857763

ABSTRACT

BACKGROUND: The authors characterized the γ-aminobutyric acid type A receptor pharmacology of the novel etomidate analog naphthalene-etomidate, a potential lead compound for the development of anesthetic-selective competitive antagonists. METHODS: The positive modulatory potencies and efficacies of etomidate and naphthalene-etomidate were defined in oocyte-expressed α1ß3γ2L γ-aminobutyric acid type A receptors using voltage clamp electrophysiology. Using the same technique, the ability of naphthalene-etomidate to reduce currents evoked by γ-aminobutyric acid alone or γ-aminobutyric acid potentiated by etomidate, propofol, pentobarbital, and diazepam was quantified. The binding affinity of naphthalene-etomidate to the transmembrane anesthetic binding sites of the γ-aminobutyric acid type A receptor was determined from its ability to inhibit receptor photoaffinity labeling by the site-selective photolabels [H]azi-etomidate and R-[H]5-allyl-1-methyl-5-(m-trifluoromethyl-diazirynylphenyl) barbituric acid. RESULTS: In contrast to etomidate, naphthalene-etomidate only weakly potentiated γ-aminobutyric acid-evoked currents and induced little direct activation even at a near-saturating aqueous concentration. It inhibited labeling of γ-aminobutyric acid type A receptors by [H]azi-etomidate and R-[H]5-allyl-1-methyl-5-(m-trifluoromethyl-diazirynylphenyl) barbituric acid with similar half-maximal inhibitory concentrations of 48 µM (95% CI, 28 to 81 µM) and 33 µM (95% CI, 20 to 54 µM). It also reduced the positive modulatory actions of anesthetics (propofol > etomidate ~ pentobarbital) but not those of γ-aminobutyric acid or diazepam. At 300 µM, naphthalene-etomidate increased the half-maximal potentiating propofol concentration from 6.0 µM (95% CI, 4.4 to 8.0 µM) to 36 µM (95% CI, 17 to 78 µM) without affecting the maximal response obtained at high propofol concentrations. CONCLUSIONS: Naphthalene-etomidate is a very low-efficacy etomidate analog that exhibits the pharmacology of an anesthetic competitive antagonist at the γ-aminobutyric acid type A receptor.


Subject(s)
Binding, Competitive/physiology , Etomidate/analogs & derivatives , Etomidate/metabolism , GABA Antagonists/metabolism , Receptors, GABA-A/metabolism , Animals , Binding, Competitive/drug effects , Dose-Response Relationship, Drug , Etomidate/pharmacology , Female , GABA Antagonists/pharmacology , Naphthalenes/chemistry , Naphthalenes/metabolism , Naphthalenes/pharmacology , Oocytes , Treatment Outcome , Xenopus laevis , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
16.
Anesthesiology ; 125(5): 943-951, 2016 11.
Article in English | MEDLINE | ID: mdl-27541316

ABSTRACT

BACKGROUND: Etomidate potently suppresses adrenocortical steroid synthesis with potentially deleterious consequences by binding to 11ß-hydroxylase and inhibiting its function. The authors hypothesized that other sedative-hypnotics currently in clinical use or under development (or their metabolites) might bind to the same site at clinically relevant concentrations. The authors tested this hypothesis by defining etomidate's affinity for this site and the potencies with which other sedative-hypnotics (and their metabolites) inhibit etomidate binding. METHODS: H-etomidate's binding to adrenal membranes from Sprague-Dawley rats was characterized with a filtration assay, and its dissociation constant was defined using saturation and homologous ligand competition approaches. Half-inhibitory concentrations of sedative-hypnotics and metabolites were determined from the reduction in specific H-etomidate binding measured in the presence of ranging sedative-hypnotic and metabolite concentrations. RESULTS: Saturation and homologous competition studies yielded H-etomidate dissociation constants of 40 and 21 nM, respectively. Half-inhibitory concentrations of etomidate and cyclopropyl methoxycarbonyl metomidate (CPMM) differed significantly (26 vs. 143 nM, respectively; P < 0.001), and those of the carboxylic acid (CA) metabolites etomidate-CA and CPMM-CA were greater than or equal to 1,000× higher than their respective parent hypnotics. The half-inhibitory concentration of dexmedetomidine was 2.2 µM, whereas those of carboetomidate, ketamine, and propofol were greater than or equal to 50 µM. CONCLUSION: Etomidate's in vitro dissociation constant for 11ß-hydroxylase closely approximates its in vivo adrenocortical half-inhibitory concentration. CPMM produces less adrenocortical suppression than etomidate not only because it is metabolized faster but also because it binds to 11ß-hydroxylase with lower affinity. Other sedative-hypnotics and metabolites bind to 11ß-hydroxylase and inhibit etomidate binding only at suprahypnotic concentrations.


Subject(s)
Adrenal Cortex/drug effects , Adrenal Cortex/metabolism , Etomidate/pharmacology , Hypnotics and Sedatives/pharmacology , Steroid 11-beta-Hydroxylase/drug effects , Steroid 11-beta-Hydroxylase/metabolism , Anesthetics, Dissociative/pharmacology , Animals , Etomidate/analogs & derivatives , Ketamine/pharmacology , Models, Animal , Propofol/pharmacology , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
17.
Anesth Analg ; 122(4): 1008-14, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26991617

ABSTRACT

BACKGROUND: Methoxycarbonyl etomidate (MOC-etomidate) and cyclopropyl methoxycarbonyl metomidate (CPMM) are rapidly metabolized "soft" etomidate analogs. CPMM's duration of hypnotic effect is context insensitive, whereas MOC-etomidate's is not. In this study, we tested the hypothesis that CPMM's effect is context insensitive because, unlike MOC-etomidate, its metabolite fails to reach physiologically important concentrations in vivo even with prolonged continuous infusion. METHODS: We compared the potencies with which MOC-etomidate and CPMM activate α1(L264T)ß3γ2 γ-aminobutyric acid type A receptors and induce loss-of-righting reflexes (i.e., produce hypnosis) in tadpoles with those of their metabolites (MOC-etomidate's carboxylic acid metabolite [MOC-ECA] and CPMM's carboxylic acid metabolite [CPMM-CA], respectively). We measured metabolite concentrations in the blood and cerebrospinal fluid of Sprague-Dawley rats on CPMM infusion and compared them with those achieved with MOC-etomidate infusion. We measured the rates with which brain tissue from Sprague-Dawley rats metabolize MOC-etomidate and CPMM. RESULTS: Both analogs and their metabolites enhanced γ-aminobutyric acid type A receptor function and induced loss-of-righting reflexes in a concentration-dependent manner. However, in these 2 assays, CPMM-CA's potency relative to its parent hypnotic was approximately 1:4900 and 1:1900, respectively, whereas MOC-ECA's was only approximately 1:415 and 1:390, respectively. With 2-hour CPMM infusions, CPMM-CA reached respective concentrations in the blood and cerebrospinal fluid that were 2 and >3 orders of magnitude lower than that which produced hypnosis. CPMM was metabolized by the brain tissue at a rate that is approximately 1/15th that of MOC-etomidate. CONCLUSIONS: Hypnotic recovery after CPMM administration is context insensitive because its metabolite does not accumulate to hypnotic levels in the central nervous system. This reflects the very large potency ratio between CPMM and CPMM-CA and the resistance of CPMM to metabolism by esterases present in the brain.


Subject(s)
Etomidate/analogs & derivatives , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/metabolism , Animals , Brain/drug effects , Brain/metabolism , Etomidate/administration & dosage , Etomidate/metabolism , Female , Larva , Rats , Rats, Sprague-Dawley , Xenopus laevis
18.
Anesthesiology ; 124(3): 651-63, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26691905

ABSTRACT

BACKGROUND: Etomidate is a highly potent anesthetic agent that is believed to produce hypnosis by enhancing γ-aminobutyric acid type A (GABAA) receptor function. The authors characterized the GABAA receptor and hypnotic potencies of etomidate analogs. The authors then used computational techniques to build statistical and graphical models that relate the potencies of these etomidate analogs to their structures to identify the specific molecular determinants of potency. METHODS: GABAA receptor potencies were defined with voltage clamp electrophysiology using α1ß3γ2 receptors harboring a channel mutation (α1[L264T]) that enhances anesthetic sensitivity (n = 36 to 60 measurements per concentration-response curve). The hypnotic potencies of etomidate analogs were defined using a loss of righting reflexes assay in Sprague Dawley rats (n = 9 to 21 measurements per dose-response curve). Three-dimensional quantitative structure-activity relationships were determined in silico using comparative molecular field analysis. RESULTS: The GABAA receptor and hypnotic potencies of etomidate and the etomidate analogs ranged by 91- and 53-fold, respectively. These potency measurements were significantly correlated (r = 0.72), but neither measurement correlated with drug hydrophobicity (r = 0.019 and 0.005, respectively). Statistically significant and predictive comparative molecular field analysis models were generated, and a pharmacophore model was built that revealed both the structural elements in etomidate analogs associated with high potency and the interactions that these elements make with the etomidate-binding site. CONCLUSIONS: There are multiple specific structural elements in etomidate and etomidate analogs that mediate GABAA receptor modulation. Modifying any one element can alter receptor potency by an order of magnitude or more.


Subject(s)
Etomidate/analogs & derivatives , Etomidate/pharmacology , GABA Modulators/pharmacology , Hypnotics and Sedatives/pharmacology , Receptors, GABA-A/physiology , Animals , Dose-Response Relationship, Drug , Female , Rats , Rats, Sprague-Dawley , Xenopus laevis
19.
J Physiol ; 593(22): 4943-61, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26378885

ABSTRACT

KEY POINTS: Most barbiturates are anaesthetics but unexpectedly a few are convulsants whose mechanism of action is poorly understood. We synthesized and characterized a novel pair of chiral barbiturates that are capable of photolabelling their binding sites on GABAA receptors. In mice the S-enantiomer is a convulsant, but the R-enantiomer is an anticonvulsant. The convulsant S-enantiomer binds solely at an inhibitory site. It is both an open state inhibitor and a resting state inhibitor. Its action is pH independent, suggesting the pyrimidine ring plays little part in binding. The inhibitory site is not enantioselective because the R-enantiomer inhibits with equal affinity. In contrast, only the anticonvulsant R-enantiomer binds to the enhancing site on open channels, causing them to stay open longer. The enhancing site is enantioselective. The in vivo actions of the convulsant S-enantiomer are accounted for by its interactions with GABAA receptors. ABSTRACT: Most barbiturates are anaesthetics but a few unexpectedly are convulsants. We recently located the anaesthetic sites on GABAA receptors (GABAA Rs) by photolabelling with an anaesthetic barbiturate. To apply the same strategy to locate the convulsant sites requires the creation and mechanistic characterization of a suitable agent. We synthesized enantiomers of a novel, photoactivable barbiturate, 1-methyl-5-propyly-5-(m-trifluoromethyldiazirinyl) phenyl barbituric acid (mTFD-MPPB). In mice, S-mTFD-MPPB acted as a convulsant, whereas R-mTFD-MPPB acted as an anticonvulsant. Using patch clamp electrophysiology and fast solution exchange on recombinant human α1 ß3 γ2L GABAA Rs expressed in HEK cells, we found that S-mTFD-MPPB inhibited GABA-induced currents, whereas R-mTFD-MPPB enhanced them. S-mTFD-MPPB caused inhibition by binding to either of two inhibitory sites on open channels with bimolecular kinetics. It also inhibited closed, resting state receptors at similar concentrations, decreasing the channel opening rate and shifting the GABA concentration-response curve to the right. R-mTFD-MPPB, like most anaesthetics, enhanced receptor gating by rapidly binding to allosteric sites on open channels, initiating a rate-limiting conformation change to stabilized open channel states. These states had slower closing rates, thus shifting the GABA concentration-response curve to the left. Under conditions when most GABAA Rs were open, an inhibitory action of R-mTFD-MPPB was revealed that had a similar IC50 to that of S-mTFD-MPPB. Thus, the inhibitory sites are not enantioselective, and the convulsant action of S-mTFD-MPPB results from its negligible affinity for the enhancing, anaesthetic sites. Interactions with these two classes of barbiturate binding sites on GABAA Rs underlie the enantiomers' different pharmacological activities in mice.


Subject(s)
Anticonvulsants/pharmacology , Convulsants/pharmacology , GABA Agents/pharmacology , Phenobarbital/analogs & derivatives , Receptors, GABA-A/metabolism , Action Potentials , Allosteric Regulation , Animals , Anticonvulsants/chemistry , Convulsants/chemistry , GABA Agents/chemistry , HEK293 Cells , Humans , Ion Channel Gating , Isomerism , Male , Mice , Phenobarbital/chemistry , Phenobarbital/pharmacology , Receptors, GABA-A/chemistry , Xenopus
20.
Anesthesiology ; 123(2): 368-76, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26035348

ABSTRACT

BACKGROUND: Cyclopropyl-methoxycarbonyl metomidate (CPMM) is a rapidly metabolized etomidate analog that is currently in clinical trials. The goal of this study is to assess CPMM's potential value as an anesthetic agent for use in patients with sepsis by defining its actions in an acute inflammatory model of sepsis. METHODS: Escherichia coli lipopolysaccharide (1 mg/kg) was injected intravenously into Sprague-Dawley rats. Thirty minutes later, CPMM, etomidate, or vehicle (n = 8 per group) was infused for 1 h. Plasma adrenocorticotropic hormone, corticosterone, and cytokine (interleukin-1ß, interleukin-6, interleukin-10, and tumor necrosis factor-α) concentrations were measured before, during, and after infusion. RESULTS: After lipopolysaccharide injection, adrenocorticotropic hormone concentrations changed similarly over time in all three groups. Compared with vehicle group rats, CPMM group rats had significantly lower corticosterone concentrations at only a single study time point during infusion and no significant differences in cytokine concentrations at any time during the study period. Compared with etomidate group rats, CPMM group rats had significantly higher corticosterone concentrations (up to nine-fold) during and after hypnotic infusion. Cytokine concentrations in CPMM group rats and vehicle group rats were not significantly different, but they were significantly lower than those in etomidate group rats. Postinfusion mortality was 40% in etomidate group rats and 0% in CPMM and vehicle group rats. CONCLUSION: Compared with etomidate, CPMM produces less adrenocortical suppression, lower plasma cytokine concentrations, and improved survival in a lipopolysaccharide inflammatory model of sepsis. These results suggest that CPMM may be a safer alternative to etomidate in patients with sepsis.


Subject(s)
Disease Models, Animal , Etomidate/analogs & derivatives , Inflammation Mediators/blood , Lipopolysaccharides/toxicity , Sepsis/blood , Sepsis/drug therapy , Animals , Etomidate/administration & dosage , Inflammation/blood , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation Mediators/antagonists & inhibitors , Infusions, Intravenous , Male , Rats , Rats, Sprague-Dawley , Sepsis/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...