1
|
Junkes L, Mendlowicz MV, Shader R, Nardi AE. Leo Sternbach and the benzodiazepines 60 years on: A revolutionary treatment for anxiety disorders. Pharmacol Res 2024; 207:107310. [PMID: 39059612 DOI: 10.1016/j.phrs.2024.107310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Affiliation(s)
- Larissa Junkes
- Institute of Psychiatry (IPUB), Federal University of Rio de Janeiro (UFRJ), Venceslau Brás Avenue, 71, Botafogo, Rio de Janeiro 22290-140, Brazil.
| | - Mauro V Mendlowicz
- Federal Fluminense University (UFF), Marquês de Paraná Avenue, 303, Downtown, Niteroi 24030-215, Brazil
| | - Richard Shader
- Tufts University, Graduate School of Biomedical Sciences, Pharmacology Program, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Antonio E Nardi
- Institute of Psychiatry (IPUB), Federal University of Rio de Janeiro (UFRJ), Venceslau Brás Avenue, 71, Botafogo, Rio de Janeiro 22290-140, Brazil
| |
Collapse
|
2
|
Dijkstra FM, Zuiker RGJA, Heuberger JAAC, Kanhai KMS, De Kam M, Duvauchelle T, Lecomte JM, Labeeuw O, Landais L, Ligneau X, Robert P, Capet M, Schwartz JC, van Gerven JMA. Administration of oxathridine, a first-in-class histamine-3 receptor partial agonist in healthy male volunteers: Central nervous system depression and pseudo-hallucinations. Br J Clin Pharmacol 2024; 90:321-335. [PMID: 37724688 DOI: 10.1111/bcp.15910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023] Open
Abstract
AIMS To characterise the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of single ascending doses of oxathridine, a first-in-class histamine-3 receptor partialagonist, in healthy male volunteers. METHODS A randomised, double-blind, placebo-controlled study including the NeuroCart, consisting of a battery of drug sensitive neurophysiological tests, was performed. Oxathridine was administered orally as an aqueous solution. After dosing, safety and NeuroCart tests (adaptive tracking [AT], body sway [BS], saccadic peak velocity [SPV], smooth pursuit [SP] eye movements, VAS according to Bond and Lader, VAS according to Bowdle [VAS B&L, Bowdle], pharmaco-electroencephalogram [pEEG], Sustained Attention to Response Task [SART]) were performed at set times. RESULTS Forty volunteers completed the study. Given doses were: 0.5, 2.5, 5, 0.25 and 1.5 mg. At 5 mg, unacceptable and unanticipated adverse events (AEs) of (orthostatic) hypotension and pseudo-hallucinations were reported. Statistically significant effects ([CI]; p-value) of 2.5 mg and 5 mg oxathridine were observed on AT ([-8.28, -1.60]; p = 0.0048), ([-8.10, -1.51]; p = 0.00530), BS ([0.6, 80.2]; p = 0.0455), ([5.9, 93.1]; p = 0.0205) and SPV ([-59.0, -15.9]; p = 0.0011), ([-43.9, -1.09]; p = 0.0399), respectively. Oxathridine 5 mg significantly increased all three VAS Bowdle subscale scores; VAS external ([0.183, 0.476]; p = <.0001), VAS internal ([0.127, 0.370]; p = 0.0001) and VAS feeling high ([0.263, 0.887]; p = 0.0006). CONCLUSION NeuroCart tests indicated central nervous system (CNS) depressant effects. Oxathridine also unexpectedly caused pseudohallucinations. Although this led to the decision to stop further development of oxathridine, these observations suggest that the H3R system could be an interesting new target for the development of novel antipsychotics.
Collapse
Affiliation(s)
- Francis M Dijkstra
- Centre for Human Drug Research, Leiden, the Netherlands
- Leiden University Medical Center, Leiden, ZA, the Netherlands
| | - Rob G J A Zuiker
- Centre for Human Drug Research, Leiden, the Netherlands
- Leiden University Medical Center, Leiden, ZA, the Netherlands
| | | | - Kawita M S Kanhai
- Centre for Human Drug Research, Leiden, the Netherlands
- Leiden University Medical Center, Leiden, ZA, the Netherlands
| | | | | | | | | | | | | | | | - Marc Capet
- Bioprojet Biotech, Saint Gregoire, France
| | | | - Joop M A van Gerven
- Centre for Human Drug Research, Leiden, the Netherlands
- Leiden University Medical Center, Leiden, ZA, the Netherlands
| |
Collapse
|
3
|
Central nervous system effects of TAK-653, an investigational alpha-amino-3-hydroxy-5-methyl-4-isoxazole receptor (AMPAR) positive allosteric modulator in healthy volunteers. Transl Psychiatry 2022; 12:408. [PMID: 36153330 PMCID: PMC9509332 DOI: 10.1038/s41398-022-02148-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/17/2022] [Accepted: 09/01/2022] [Indexed: 11/24/2022] Open
Abstract
TAK-653 is a novel AMPA receptor positive allosteric modulator in clinical development for the treatment of major depressive disorder (MDD). This study aimed to measure the functional pharmacodynamic central nervous system (CNS) effects of TAK-653. A randomised, double-blind, placebo-controlled, three-way crossover (placebo, TAK-653 0.5 mg and 6 mg) study with 24 healthy volunteers was performed. NeuroCart tests consisting of body sway (BS), saccadic peak velocity (SPV), smooth pursuit eye movements (SP), adaptive tracking (AT), Bowdle and Bond and Lader Visual Analogue Scales (B-VAS and BL-VAS) and Stroop test were performed pre-dose and 3.5 and 4 h post-dose. Data were analysed using a mixed model analysis of covariance with baseline as covariate. It was found that TAK-653 did not affect BS and subjective drug effects as measured by B-VAS and BL-VAS at either dose level. TAK-653 0.5 mg increased SPV (degrees/second) (19.49 [5.98, 32.99], P = 0.02) and affected Stroop difference in reaction time between correct congruent and correct incongruent answers and number of correct responses in incongruent trials (22.0 [4.0, 40.0], P = 0.05 and -0.3 [-0.5, -0.1], P = 0.02, respectively). TAK-653 6 mg improved AT (%) (1.68 [0.51, 2.84], P = 0.02) and increased SPV (degrees/s) (15.40 [1.91, 28.90], P = 0.06) and SP (%) (2.32 [0.37, 4.27], P = 0.05). Based on these findings it can be concluded that TAK-653 demonstrated a psychostimulant-like pharmacodynamic profile on the NeuroCart consistent with previously reported increase of cortical excitability following Transcranial Magnetic Stimulation (TMS) of the human motor cortex.
Collapse
|
4
|
Ito M, Spence A, Blauwet MB, Heo N, Goldwater R, Maruff P, Marek GJ. A phase 1 study to assess potential interaction between ASP8062 and alcohol in healthy adult subjects. J Psychopharmacol 2022; 36:756-767. [PMID: 34994232 DOI: 10.1177/02698811211058967] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND ASP8062 is a novel orally active GABAB receptor positive allosteric modulator in clinical development for the treatment of alcohol use disorder (AUD) and opioid use disorder (OUD). AIMS This study assessed the potential pharmacokinetic/pharmacodynamic interaction between ASP8062 and alcohol under single-dose conditions in healthy adults. METHODS A double-blind, placebo-controlled, crossover phase 1 study was conducted in which 20 subjects were randomly assigned to four treatment sequences (ASP8062 + alcohol; ASP8062 + placebo alcohol; placebo + alcohol; placebo + placebo alcohol) each consisting of four treatment periods, separated by washout periods of at least 14 days. An analysis of variance was used to assess pharmacokinetic interaction and a mixed-effects analysis of covariance was used to assess pharmacodynamic interaction. RESULTS/OUTCOMES After administration of alcohol, a mild to minimal increase in plasma exposure (AUCinf and Cmax) of ASP8062 was observed, but tmax and t½ for ASP8062 remained unchanged after administration of alcohol. In contrast, ASP8062 did not affect the AUClast and Cmax of ethanol. No clinically relevant differences in cognition measurements were observed with ASP8062 compared with placebo, but there were expected impairments in psychomotor and executive function with alcohol alone. ASP8062 in combination with alcohol resulted in worse scores in cognition measurements than alcohol alone, but this potentiation was not consistent. ASP8062 administered alone was safe and well-tolerated and safety findings in subjects administered alcohol alone were not augmented when ASP8062 was administered in combination with alcohol. CONCLUSION/INTERPRETATION The data support further clinical studies investigating ASP8062 in patients with AUD.
Collapse
Affiliation(s)
- Mototsugu Ito
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Anna Spence
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | | | - Nakyo Heo
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | | | | | - Gerard J Marek
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| |
Collapse
|
5
|
Faßbender K, Bey K, Lippold JV, Aslan B, Hurlemann R, Ettinger U. GABAergic modulation of performance in response inhibition and interference control tasks. J Psychopharmacol 2021; 35:1496-1509. [PMID: 34278874 DOI: 10.1177/02698811211032440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Inhibitory control is a crucial executive function with high relevance to mental and physical well-being. However, there are still unanswered questions regarding its neural mechanisms, including the role of the major inhibitory neurotransmitter, γ-aminobutyric acid (GABA). AIMS This study examined the effects of lorazepam (0.5 mg and 1 mg), a positive allosteric modulator at the GABAA receptor, on response inhibition and interference control. We also explored the heterogeneity of inhibitory control and calculated delta plots to explore whether lorazepam affects the gradual build-up of inhibition and activation over time. METHODS N = 50 healthy participants performed antisaccade, Eriksen flanker and Simon tasks in a within-subjects, placebo-controlled, double-blind randomized design. RESULTS Lorazepam increased reaction time (RT) and error rates dose dependently in all tasks (p ⩽ 0.005). In the antisaccade and Simon tasks, lorazepam increased congruency effects for error rate (p ⩽ 0.029) but not RT (p ⩾ 0.587). In the Eriksen flanker task, both congruency effects were increased by the drug (p ⩽ 0.031). Delta plots did not reflect drug-induced changes in inhibition and activation over time. Delta plots for RT in the Simon task were negative-going, as expected, whereas those for the antisaccade and flanker tasks were positive-going. CONCLUSIONS This study provides evidence for GABAergic involvement in performance on response inhibition and interference control tasks. Furthermore, our findings highlight the diversity of the broader construct of inhibitory control while also pointing out similarities between different inhibitory control tasks. In contrast to RT and error rates, the cognitive processes indexed by delta plots may not be sensitive to GABAergic modulation.
Collapse
Affiliation(s)
- Kaja Faßbender
- Department of Psychology, University of Bonn, Bonn, Germany
| | - Katharina Bey
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | | | - Behrem Aslan
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - René Hurlemann
- Department of Psychiatry, School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| | | |
Collapse
|
6
|
White JP, Schembri A, Edgar CJ, Lim YY, Masters CL, Maruff P. A Paradox in Digital Memory Assessment: Increased Sensitivity With Reduced Difficulty. Front Digit Health 2021; 3:780303. [PMID: 34881380 PMCID: PMC8645569 DOI: 10.3389/fdgth.2021.780303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/01/2021] [Indexed: 12/16/2022] Open
Abstract
The One Card Learning Test (OCL80) from the Cogstate Brief Battery-a digital cognitive test used both in-person and remotely in clinical trials and in healthcare contexts to inform health decisions-has shown high sensitivity to changes in memory in early Alzheimer's disease (AD). However, recent studies suggest that OCL sensitivity to memory impairment in symptomatic AD is not as strong as that for other standardized assessments of memory. This study aimed to improve the sensitivity of the OCL80 to AD-related memory impairment by reducing the test difficultly (i.e., OCL48). Experiment 1 showed performance in healthy adults improved on the OCL48 while the pattern separation operations that constrain performance on the OCL80 were retained. Experiment 2 showed repeated administration of the OCL48 at short retest intervals did not induce ceiling or practice effects. Experiment 3 showed that the sensitivity of the OCL48 to AD-related memory impairment (Glass's Δ = 3.11) was much greater than the sensitivity of the OCL80 (Glass's Δ = 1.94). Experiment 4 used data from a large group of cognitively normal older adults to calibrate performance scores between the OCL80 and OCL48 using equipercentile equating. Together these results showed the OCL48 to be a valid and reliable test of learning with greater sensitivity to memory impairment in AD than the OCL80.
Collapse
Affiliation(s)
| | | | | | - Yen Ying Lim
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Colin L. Masters
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul Maruff
- Cogstate Ltd, Melbourne, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
7
|
Chen X, Sang N, Song K, Zhong W, Wang H, Jiang J, Huang Y, Hu P. Psychomotor Recovery Following Remimazolam-induced Sedation and the Effectiveness of Flumazenil as an Antidote. Clin Ther 2020; 42:614-624. [PMID: 32178858 DOI: 10.1016/j.clinthera.2020.02.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 01/31/2020] [Accepted: 02/08/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE Remimazolam tosylate (HR-7056) is a novel ester-type benzodiazepine with ultrafast onset of effect. The compound is being developed for sedation induction and maintenance during anesthesia. It was approved for procedural anesthesia in December 2019 by the National Medical Products Administration of China. Previous studies have reported on remimazolam's effects on consciousness and cognition. Although the time to full psychomotor recovery after remimazolam-mediated sedation is critical for decisions regarding hospital discharge, relevant clinical evidence is still lacking. This study investigated the residual psychomotor effects of remimazolam and their recovery from sedating treatment in 2 simulated clinical settings: (1) single-dose administration for sedation initiation; and (2) constant rate infusion for sedation maintenance. METHODS A single-ascending-dose, parallel-group, midazolam-controlled study and a 2-way crossover study evaluating the reversal effect of flumazenil versus placebo after a 2-h constant rate infusion were conducted with HR-7056 in 87 Chinese healthy volunteers; the studies used a double-blind, randomized trial design. A battery of psychomotor tests was administered before dosing and several times postdose over 4-6 h. Pharmacokinetic, sedation, and safety assessments were performed throughout the studies. FINDINGS After bolus infusion, the Bispectral Index score decreased in a concentration-dependent manner with HR-7056, accompanied by a sharp drop of Modified Observer's Assessment of Alertness/Sedation score. The recovery of consciousness was much faster with HR-7056 than with midazolam. During the constant rate infusion, the Bispectral Index score was maintained between 40 and 60 with an average plasma remimazolam concentration of ~1000 ng/mL. Subjects' performance in saccadic and smooth pursuit eye movement, body sway, test of choice reaction time, and word recall was significantly impaired after single-dose midazolam and after constant rate infusion of remimazolam. The end-of-infusion injection of flumazenil shortened the median time to full alertness to 3.5 min and effectively reversed psychomotor and cardiovascular dysfunction. IMPLICATIONS The study results showed quicker psychomotor recovery from sedation in the remimazolam-treated group. The moderate and short-lasting residual effect of remimazolam after 2-h conscious sedation proposes a need for psychomotor assessment(s) before hospital discharge. ClinicalTrials.gov identifiers: NCT01970072 and NCT03444480.
Collapse
Affiliation(s)
- Xia Chen
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Beijing, China; Clinical Trial Center, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Nuoer Sang
- Department of Anesthesiology, Peking Union Medical College Hospital, Beijing, China
| | - Kaicheng Song
- Department of Anesthesiology, Peking Union Medical College Hospital, Beijing, China
| | - Wen Zhong
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Beijing, China
| | - Hongyun Wang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Beijing, China
| | - Ji Jiang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Beijing, China
| | - Yuguang Huang
- Department of Anesthesiology, Peking Union Medical College Hospital, Beijing, China
| | - Pei Hu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|
8
|
Wang W, Wang Y, Zhao W, Zhong J, Wang Y, Chen X. Pharmacokinetics, pharmacodynamics, safety, tolerability, and mass balance of single and continuous intravenous infusion of SPT-07A in healthy volunteers. Eur J Clin Pharmacol 2020; 76:785-793. [PMID: 32147750 DOI: 10.1007/s00228-020-02851-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/26/2020] [Indexed: 11/30/2022]
Abstract
PURPOSE SPT-07A is an intravenous injection of (+)-2-borneol being developed for the treatment of acute ischemic stroke. This study aimed to investigate the pharmacokinetics, pharmacodynamics, safety, tolerability, and mass balance of SPT-07A after sequentially administered single and multiple infusions of SPT-07A at 10 mg, 20 mg, or 40 mg. METHODS This phase I, double-blind, randomized, placebo-controlled, dose-escalation study was conducted in 36 Chinese healthy volunteers. Each cohort enrolled 12 eligible subjects, who were 9:3 randomized to receive SPT-07A or matching placebo during the two study occasions, that is, an initial single-dose occasion followed by a 7-day multiple-dose occasion with a dosing interval of 12 h. Pharmacokinetic, pharmacodynamic assessments regarding effects on the central nervous system (CNS) were performed pre-dose and several times post-dose. Safety and tolerability were evaluated throughout the study for each cohort. RESULTS Following single intravenous (i.v.) administration of 10 mg to 40 mg SPT-07A, the plasma SPT-07A concentration reached its peak by the end of infusion. Thereafter, the plasma concentration declined in a multiphase exponential manner with an average terminal elimination half-life of 3.85 to 8.93 h. The exposure parameters of SPT-07A increased dose proportionally. Steady state of SPT-07A was reached after 12-hourly i.v. administrations for 4 days with minimal accumulations. No significant difference of change-from-baseline was observed in the pharmacodynamic measurements between each of the three SPT-07A-treated groups and the placebo group. A total of 41 adverse events (AEs) were reported in 77.8% subjects at 10 mg (7/9), 20 mg (7/9), and 40 mg (7/9), respectively. The AE incidence in placebo group was also 77.8% (7/9). All AEs were mild or moderate in severity and self-limited. SPT-07A was mainly excreted in human urine in glucuronic acid conjugate forms. The total urine recovery rate approximated 84.69% of the administered dose. CONCLUSIONS SPT-07A was safe and well tolerated after single and multiple intravenous administrations of SPT-07A in the range of 10 mg to 40 mg. SPT-07A presented linear pharmacokinetics in human. Based on plasma exposure, the doses of 10-40 mg twice daily resulted in exposure levels comparable with those obtained at doses demonstrating potential efficacy on AIS animal models and were thus recommended as therapeutic exploratory doses in the phase II clinical trial.
Collapse
Affiliation(s)
- Weicong Wang
- Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Yan Wang
- Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Weiwei Zhao
- Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Jingbo Zhong
- Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Yongjun Wang
- Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Xia Chen
- Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China.
| |
Collapse
|
9
|
Maramai S, Benchekroun M, Ward SE, Atack JR. Subtype Selective γ-Aminobutyric Acid Type A Receptor (GABAAR) Modulators Acting at the Benzodiazepine Binding Site: An Update. J Med Chem 2019; 63:3425-3446. [DOI: 10.1021/acs.jmedchem.9b01312] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Samuele Maramai
- Sussex Drug Discovery Centre, University of Sussex, Brighton BN1 9QJ, U.K
| | - Mohamed Benchekroun
- Sussex Drug Discovery Centre, University of Sussex, Brighton BN1 9QJ, U.K
- Équipe de Chimie Moléculaire, Laboratoire de Génomique Bioinformatique et Chimie Moléculaire, GBCM, EA7528, Conservatoire National des Arts et Métiers, 2 rue Conté, 75003 Paris, France
| | - Simon E. Ward
- Medicines Discovery Institute, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K
| | - John R. Atack
- Medicines Discovery Institute, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K
| |
Collapse
|
10
|
Chen X, van Gerven J, Cohen A, Jacobs G. Human pharmacology of positive GABA-A subtype-selective receptor modulators for the treatment of anxiety. Acta Pharmacol Sin 2019; 40:571-582. [PMID: 30518829 DOI: 10.1038/s41401-018-0185-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022] Open
Abstract
Anxiety disorders arise from disruptions among the highly interconnected circuits that normally serve to process the streams of potentially threatening stimuli. The resulting imbalance among these circuits can cause a fundamental misinterpretation of neural sensory information as threatening and can lead to the inappropriate emotional and behavioral responses observed in anxiety disorders. There is considerable preclinical evidence that the GABAergic system, in general, and its α2- and/or α5-subunit-containing GABA(A) receptor subtypes, in particular, are involved in the pathophysiology of anxiety disorders. However, the clinical efficacy of GABA-A α2-selective agonists for the treatment of anxiety disorders has not been unequivocally demonstrated. In this review, we present several human pharmacological studies that have been performed with the aim of identifying the pharmacologically active doses/exposure levels of several GABA-A subtype-selective novel compounds with potential anxiolytic effects. The pharmacological selectivity of novel α2-subtype-selective GABA(A) receptor partial agonists has been demonstrated by their distinct effect profiles on the neurophysiological and neuropsychological measurements that reflect the functions of multiple CNS domains compared with those of benzodiazepines, which are nonselective, full GABA(A) agonists. Normalizing the undesired pharmacodynamic side effects against the desired on-target effects on the saccadic peak velocity is a useful approach for presenting the pharmacological features of GABA(A)-ergic modulators. Moreover, combining the anxiogenic symptom provocation paradigm with validated neurophysiological and neuropsychological biomarkers may provide further construct validity for the clinical effects of novel anxiolytic agents. In addition, the observed drug effects on serum prolactin levels support the use of serum prolactin levels as a complementary neuroendocrine biomarker to further validate the pharmacodynamic measurements used during the clinical pharmacological study of novel anxiolytic agents.
Collapse
|
11
|
An 8-Week, Randomized, Phase 2, Double-Blind, Sequential Parallel-Group Comparison Study of Two Dose Levels of the GABAA Positive Allosteric Modulator PF-06372865 Compared With Placebo as an Adjunctive Treatment in Outpatients With Inadequate Response to Standard of Care for Generalized Anxiety Disorder. J Clin Psychopharmacol 2019; 39:20-27. [PMID: 30531477 DOI: 10.1097/jcp.0000000000000997] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Generalized anxiety disorder (GAD) is a common psychiatric disorder, but many patients experience only partial relief of symptoms with existing therapies. Benzodiazepines are effective in many cases but are limited by a number of significant adverse effects. PF-06372865 is a subtype-selective gamma-aminobutyric acid A (GABAA)-positive allosteric modulator lacking in functional activity at alpha 1-containing receptors that are believed to mediate many of these adverse effects. METHODS PF-06372865 was evaluated as an adjunct to current GAD treatment in a double-blind, placebo-controlled, sequential parallel comparison study in patients with GAD who showed an incomplete response to current standard-of-care pharmacotherapy. A total of 90 subjects (of the planned 384) were randomized into the study before the decision to terminate the study. Two doses of PF-06372865 (2.5 mg twice daily and 7.5 mg twice daily) were compared with placebo. RESULTS Neither dose of PF-06372865 differentiated from placebo on week 4 Hamilton Anxiety Inventory total (primary end point) or on the Sheehan Disability Scale total score (secondary end point). Adverse events including dizziness, headache, and somnolence were observed, and the 7.5 mg dose demonstrated some impairment on the Digit Symbol Substitution test and the Epworth Sleepiness Scale relative to placebo and the 2.5 mg dose. CONCLUSIONS Factors contributing to the negative results include the limited sample size and failure to explore a broader range of doses.
Collapse
|
12
|
Payghan PV, Nath Roy S, Bhattacharyya D, Ghoshal N. Cross-talk between allosteric and orthosteric binding sites of γ-amino butyric acid type A receptors (GABAA-Rs): A computational study revealing the structural basis of selectivity. J Biomol Struct Dyn 2019; 37:3065-3080. [DOI: 10.1080/07391102.2018.1508367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Pavan V. Payghan
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | | | | | - Nanda Ghoshal
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
13
|
Ettinger U, Meyhöfer I, Mehta MA, Kumari V, Corr PJ, Williams SC, Perkins AM. Effects of lorazepam on saccadic eye movements: the role of sex, task characteristics and baseline traits. J Psychopharmacol 2018; 32:678-690. [PMID: 29783905 DOI: 10.1177/0269881118772450] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Saccadic eye movements are controlled by a network of parietal, frontal, striatal, cerebellar and brainstem regions. The saccadic peak velocity is an established biomarker of benzodiazepine effects, with benzodiazepines reliably reducing the peak velocity. AIMS In this study, we aimed to replicate the effects of benzodiazepines on peak velocity and we investigated effects on previously less studied measures of saccades. We also explored the roles of sex, task characteristics and the baseline variables age, intelligence and trait anxiety in these effects. METHOD Healthy adults ( N = 34) performed a horizontal step prosaccade task under 1 mg lorazepam, 2 mg lorazepam and placebo in a double-blind, within-subjects design. RESULTS We replicated the dose-dependent reduction in peak velocity with lorazepam and showed that this effect is stronger for saccades to targets at smaller eccentricities. We also demonstrated that this effect is independent of sex and other baseline variables. Lorazepam effects were widespread, however, occurring on mean and variability measures of most saccadic variables. Additionally, there were sex-dependent lorazepam effects on spatial consistency of saccades, indicating more adverse effects in females. CONCLUSIONS We conclude that saccadic peak velocity is a sensitive and robust biomarker of benzodiazepine effects. However, lorazepam has pronounced effects also on other parameters of horizontal saccades. Sex-dependent drug effects on spatial consistency may reflect cerebellar mechanisms, given the role of the cerebellum in saccadic spatial accuracy.
Collapse
Affiliation(s)
| | - Inga Meyhöfer
- 1 Department of Psychology, University of Bonn, Germany
| | - Mitul A Mehta
- 2 Department of Neuroimaging, King's College London, UK
| | - Veena Kumari
- 3 Centre for Cognitive Neuroscience, Brunel University London, Uxbridge, UK
| | - Philip J Corr
- 4 Department of Psychology, City University of London, UK
| | | | - Adam M Perkins
- 5 Department of Psychological Medicine, King's College London, UK
| |
Collapse
|
14
|
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: 2.0] [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
|
15
|
Jucaite A, Cselényi Z, Lappalainen J, McCarthy DJ, Lee CM, Nyberg S, Varnäs K, Stenkrona P, Halldin C, Cross A, Farde L. GABA A receptor occupancy by subtype selective GABA Aα2,3 modulators: PET studies in humans. Psychopharmacology (Berl) 2017; 234:707-716. [PMID: 28013354 PMCID: PMC5263201 DOI: 10.1007/s00213-016-4506-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 12/08/2016] [Indexed: 01/04/2023]
Abstract
RATIONALE Sedation, dependence, and abuse liability limit the use of non-selective γ-aminobutyric acid (GABAA) receptor positive modulators for the treatment of anxiety. AZD7325 and AZD6280 are novel, subtype-selective GABAAα2,3 receptor positive modulators with limited sedative effects. OBJECTIVES The current study aimed to confirm target engagement at GABAA receptors by AZD7325 and AZD6280 in humans and to determine the relationship between exposure, GABAA receptor occupancy, and tolerability. METHOD Two PET studies, using high-resolution research tomography (HRRT) and the radioligand [11C]flumazenil, were performed in 12 subjects at baseline and after administration of single oral doses of AZD7325 (0.2 to 30 mg) and AZD6280 (5 to 40 mg). PET images were analyzed using a simplified reference tissue model, and regional binding potentials (BPND) were obtained. The relationship between plasma concentration of AZD7325 or AZD6280 and GABAA receptor occupancy was described by hyperbolic function, and K i,plasma (plasma concentration required for 50% receptor occupancy) was estimated. Assessments of safety and tolerability included recording of adverse events, vital signs, electrocardiogram, and laboratory tests. RESULTS The [11C]flumazenil binding was reduced in a dose-dependent, saturable manner by both agents. Maximum receptor occupancy could be reached for both compounds without causing sedation or cognitive impairment. The K i,plasma estimates for AZD7325 and AZD6280 were 15 and 440 nmol/l, respectively. CONCLUSION High GABAA receptor occupancy by AZD7325 and AZD6280 could be reached without clear sedative effects.
Collapse
Affiliation(s)
- Aurelija Jucaite
- Department of Clinical Neuroscience, AstraZeneca PET Center, Karolinska Institutet, R5:02, SE-17176, Stockholm, Sweden. .,Department of Clinical Neuroscience, PET Centre, Karolinska Institutet, Stockholm, Sweden.
| | - Zsolt Cselényi
- 0000 0004 1937 0626grid.4714.6Department of Clinical Neuroscience, AstraZeneca PET Center, Karolinska Institutet, R5:02, SE-17176 Stockholm, Sweden ,0000 0004 1937 0626grid.4714.6Department of Clinical Neuroscience, PET Centre, Karolinska Institutet, Stockholm, Sweden
| | - Jaakko Lappalainen
- AstraZeneca Neuroscience Innovative Medicines, Cambridge, MA USA ,Marinus Pharmaceuticals, Radnor, PA USA
| | - Dennis J. McCarthy
- 0000 0001 1519 6403grid.418151.8AstraZeneca R&D, Södertälje, Sweden ,Independent Consultant, Newark, DE USA
| | - Chi-Ming Lee
- 0000 0001 1519 6403grid.418151.8AstraZeneca R&D, Södertälje, Sweden ,Ever East Consultants Limited, Hong Kong, People’s Republic of China
| | - Svante Nyberg
- 0000 0001 1519 6403grid.418151.8AstraZeneca R&D, Södertälje, Sweden ,0000 0000 9241 5705grid.24381.3cDepartment of Psychiatry, Karolinska University Hospital (Huddinge), Stockholm, Sweden
| | - Katarina Varnäs
- 0000 0004 1937 0626grid.4714.6Department of Clinical Neuroscience, PET Centre, Karolinska Institutet, Stockholm, Sweden
| | - Per Stenkrona
- 0000 0004 1937 0626grid.4714.6Department of Clinical Neuroscience, PET Centre, Karolinska Institutet, Stockholm, Sweden
| | - Christer Halldin
- 0000 0004 1937 0626grid.4714.6Department of Clinical Neuroscience, PET Centre, Karolinska Institutet, Stockholm, Sweden
| | - Alan Cross
- AstraZeneca Neuroscience Innovative Medicines, Cambridge, MA USA
| | - Lars Farde
- 0000 0004 1937 0626grid.4714.6Department of Clinical Neuroscience, AstraZeneca PET Center, Karolinska Institutet, R5:02, SE-17176 Stockholm, Sweden ,0000 0004 1937 0626grid.4714.6Department of Clinical Neuroscience, PET Centre, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
16
|
Zuiker RGJA, Chen X, Østerberg O, Mirza NR, Muglia P, de Kam M, Klaassen ES, van Gerven JMA. NS11821, a partial subtype-selective GABAA agonist, elicits selective effects on the central nervous system in randomized controlled trial with healthy subjects. J Psychopharmacol 2016; 30:253-62. [PMID: 26655084 DOI: 10.1177/0269881115620435] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
NS11821 is a partial GABAA agonist with relatively dominant α2,3 and α5 subtype efficacy but negligible α1 agonism. This first-in-human study was performed in healthy male subjects using a single-dose, parallel, double blind, placebo-controlled, randomized, dose-escalation study design. In total six cohorts (N=48) were enrolled. The eight subjects of each cohort received NS11821 (10 mg, 30 mg, 75 mg, 150 mg, 300 mg or 600 mg) or placebo in a 6:2 ratio. At low dose levels, NS11821 had a relatively low exposure and a more-than-proportional increase of the area under the curve and maximum plasma concentrations, probably due to poor solubility. Saccadic peak velocity decreased in a dose-related manner while limited impairments were seen on body sway and the visual analogue scale for alertness. The most common adverse events were somnolence and dizziness, which were more prominent with the higher doses. Although no positive control was used in this study, the results were compared post hoc with a Centre for Human Drug Research dataset for lorazepam 2 mg. The maximum saccadic peak velocity effects seemed comparable to the typical effects of lorazepam, whereas the other central nervous system effects were smaller. These results support the pharmacological selectivity of NS11821 and show that pharmacodynamic effective doses of NS11821 were safe and well tolerated in healthy subjects.
Collapse
Affiliation(s)
| | - Xia Chen
- Centre for Human Drug Research (CHDR), Leiden, the Netherlands Clinical Pharmacological Research Centre (CPRC), Peking Union Medical College Hospital, Beijing, PR China
| | | | | | | | - Marieke de Kam
- Centre for Human Drug Research (CHDR), Leiden, the Netherlands
| | | | | |
Collapse
|
17
|
MacDonald LA, Minahan CL. Indices of cognitive function measured in rugby union players using a computer-based test battery. J Sports Sci 2016; 34:1669-74. [DOI: 10.1080/02640414.2015.1132003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|