Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Journal of Pharmacology and Experimental Therapeutics
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • My Cart
Journal of Pharmacology and Experimental Therapeutics

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Visit jpet on Facebook
  • Follow jpet on Twitter
  • Follow jpet on LinkedIn
Research ArticleDrug Discovery and Translational Medicine

The Use of Physiology-Based Pharmacokinetic and Pharmacodynamic Modeling in the Discovery of the Dual Orexin Receptor Antagonist ACT-541468

Alexander Treiber, Ruben de Kanter, Catherine Roch, John Gatfield, Christoph Boss, Markus von Raumer, Benno Schindelholz, Clemens Muehlan, Joop van Gerven and Francois Jenck
Journal of Pharmacology and Experimental Therapeutics September 2017, 362 (3) 489-503; DOI: https://doi.org/10.1124/jpet.117.241596
Alexander Treiber
Departments of Preclinical Drug Metabolism and Pharmacokinetics (A.T., R.d.K.), Preclinical Pharmacology (C.R., F.J.), Biology (J.G.), Chemistry (C.B.), Clinical Pharmacology (C.M.), and Preclinical Development (M.v.R., B.S.), Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland; Center for Human Drug Research, Leiden, The Netherlands (J.v.G.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ruben de Kanter
Departments of Preclinical Drug Metabolism and Pharmacokinetics (A.T., R.d.K.), Preclinical Pharmacology (C.R., F.J.), Biology (J.G.), Chemistry (C.B.), Clinical Pharmacology (C.M.), and Preclinical Development (M.v.R., B.S.), Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland; Center for Human Drug Research, Leiden, The Netherlands (J.v.G.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Catherine Roch
Departments of Preclinical Drug Metabolism and Pharmacokinetics (A.T., R.d.K.), Preclinical Pharmacology (C.R., F.J.), Biology (J.G.), Chemistry (C.B.), Clinical Pharmacology (C.M.), and Preclinical Development (M.v.R., B.S.), Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland; Center for Human Drug Research, Leiden, The Netherlands (J.v.G.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
John Gatfield
Departments of Preclinical Drug Metabolism and Pharmacokinetics (A.T., R.d.K.), Preclinical Pharmacology (C.R., F.J.), Biology (J.G.), Chemistry (C.B.), Clinical Pharmacology (C.M.), and Preclinical Development (M.v.R., B.S.), Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland; Center for Human Drug Research, Leiden, The Netherlands (J.v.G.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Christoph Boss
Departments of Preclinical Drug Metabolism and Pharmacokinetics (A.T., R.d.K.), Preclinical Pharmacology (C.R., F.J.), Biology (J.G.), Chemistry (C.B.), Clinical Pharmacology (C.M.), and Preclinical Development (M.v.R., B.S.), Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland; Center for Human Drug Research, Leiden, The Netherlands (J.v.G.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Markus von Raumer
Departments of Preclinical Drug Metabolism and Pharmacokinetics (A.T., R.d.K.), Preclinical Pharmacology (C.R., F.J.), Biology (J.G.), Chemistry (C.B.), Clinical Pharmacology (C.M.), and Preclinical Development (M.v.R., B.S.), Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland; Center for Human Drug Research, Leiden, The Netherlands (J.v.G.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Benno Schindelholz
Departments of Preclinical Drug Metabolism and Pharmacokinetics (A.T., R.d.K.), Preclinical Pharmacology (C.R., F.J.), Biology (J.G.), Chemistry (C.B.), Clinical Pharmacology (C.M.), and Preclinical Development (M.v.R., B.S.), Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland; Center for Human Drug Research, Leiden, The Netherlands (J.v.G.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Clemens Muehlan
Departments of Preclinical Drug Metabolism and Pharmacokinetics (A.T., R.d.K.), Preclinical Pharmacology (C.R., F.J.), Biology (J.G.), Chemistry (C.B.), Clinical Pharmacology (C.M.), and Preclinical Development (M.v.R., B.S.), Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland; Center for Human Drug Research, Leiden, The Netherlands (J.v.G.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Joop van Gerven
Departments of Preclinical Drug Metabolism and Pharmacokinetics (A.T., R.d.K.), Preclinical Pharmacology (C.R., F.J.), Biology (J.G.), Chemistry (C.B.), Clinical Pharmacology (C.M.), and Preclinical Development (M.v.R., B.S.), Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland; Center for Human Drug Research, Leiden, The Netherlands (J.v.G.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Francois Jenck
Departments of Preclinical Drug Metabolism and Pharmacokinetics (A.T., R.d.K.), Preclinical Pharmacology (C.R., F.J.), Biology (J.G.), Chemistry (C.B.), Clinical Pharmacology (C.M.), and Preclinical Development (M.v.R., B.S.), Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland; Center for Human Drug Research, Leiden, The Netherlands (J.v.G.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF + SI
  • PDF
Loading

Abstract

The identification of new sleep drugs poses particular challenges in drug discovery owing to disease-specific requirements such as rapid onset of action, sleep maintenance throughout major parts of the night, and absence of residual next-day effects. Robust tools to estimate drug levels in human brain are therefore key for a successful discovery program. Animal models constitute an appropriate choice for drugs without species differences in receptor pharmacology or pharmacokinetics. Translation to man becomes more challenging when interspecies differences are prominent. This report describes the discovery of the dual orexin receptor 1 and 2 (OX1 and OX2) antagonist ACT-541468 out of a class of structurally related compounds, by use of physiology-based pharmacokinetic and pharmacodynamic (PBPK-PD) modeling applied early in drug discovery. Although all drug candidates exhibited similar target receptor potencies and efficacy in a rat sleep model, they exhibited large interspecies differences in key factors determining their pharmacokinetic profile. Human PK models were built on the basis of in vitro metabolism and physicochemical data and were then used to predict the time course of OX2 receptor occupancy in brain. An active ACT-541468 dose of 25 mg was estimated on the basis of OX2 receptor occupancy thresholds of about 65% derived from clinical data for two other orexin antagonists, almorexant and suvorexant. Modeling predictions for ACT-541468 in man were largely confirmed in a single-ascending dose trial in healthy subjects. PBPK-PD modeling applied early in drug discovery, therefore, has great potential to assist in the identification of drug molecules when specific pharmacokinetic and pharmacodynamic requirements need to be met.

Introduction

Insomnia, characterized by difficulties with sleep onset and/or sleep maintenance, affects 10–15% of the population chronically, with additional 15–20% suffering from it occasionally. Insomnia has a wide range of effects on quality of life and is associated with increased accident risk and chronic health problems (Zammit, 2007). Historically, insomnia treatments have targeted GABA-A, histamine, serotonin, or melatonin receptors. GABA-A receptor modulators such as zolpidem, which currently dominate the market, tend to increase non–rapid eye movement (non-REM) sleep and decrease REM sleep compared with normal sleep architecture (Bettica et al., 2012b). This may be the reason for the observed decrease in cognitive performance and locomotor skills when walking during night-time awakening (Otmani et al., 2008; Frey et al., 2011), the latter being of particular concern for the elderly (Glass et al., 2005).

The orexin system was discovered in the late 1990s (de Lecea et al., 1998; Sakurai, 1999) and is composed of two peptides, orexin A (OxA) and orexin B (OxB), produced in a small number of neurons in the lateral hypothalamus, and two G protein-coupled receptors, orexin-1 (OX1) and orexin-2 (OX2), widely expressed throughout the brain. Orexin deficiency has been linked to human narcolepsy/cataplexy, a neurologic disorder in which patients suffer from an uncontrolled sleep-wake cycle (Peyron et al., 2000; Thannickal et al., 2000). This indicates that the orexin system plays an essential role in promoting alertness and maintaining wakefulness under motivational circumstances. It supports behavioral and physiologic adaptation to relevant internal and external environmental stimuli, such as physiologic need states, exposure to threats, or reward opportunities (Sakurai, 2007; Carter et al., 2009; Tsujino and Sakurai, 2009; Mahler et al., 2014).

With the discovery of potent low-molecular-weight compounds, orexin receptor antagonism has emerged as a novel concept for the treatment of insomnia disorders. Unlike classic hypnotics targeting GABA-A, histamine, or serotonin receptors, orexin receptor antagonists promote sleep in a distinct manner, as they maintain a natural sleep architecture and do not impair cognitive function or locomotor skills (Hoever et al., 2012b; Ramirez et al., 2013; Tannenbaum et al., 2016).

Both, dual orexin receptor antagonists (DORAs) including almorexant (Hoever et al., 2012b), suvorexant (Herring et al., 2012), filorexant (Connor et al., 2016), lemborexant (Yoshida et al., 2015), and SB-649868 (Bettica et al., 2012a), as well as the OX2-selective antagonist seltorexant have been tested in insomnia patients (Bonaventure et al., 2015a). Polysomnography in animals and healthy human subjects suggest a qualitatively comparable sleep-promoting profile for both, DORAs and OX2-selective compounds, although relative impact on REM versus non-REM sleep may differ (Dugovic et al., 2014; Jacobson et al., 2017). Comparative studies with DORAs and OX2-selective compounds indicate that higher OX2 receptor occupancies are required by OX2-selective compounds (Mieda et al., 2011; Gotter et al., 2016), as recently shown with the DORA filorexant and the OX2-selective MK-1064. As OX1-selective antagonists alone lack sleep-promoting efficacy (Gozzi et al., 2011), the overall data suggest a complementary role of OX1 antagonism in the sleep-promoting efficacy of DORAs. OX1 receptors also appear to play an essential role in anxiety states (Merlo Pich and Melotto, 2014), and OX1-selective antagonists additionally attenuate behavioral and cardiovascular responses to stress without altering baseline motor or autonomic functions (Gozzi et al., 2011; Johnson et al., 2012; Bonaventure et al., 2015b).

In 2014, suvorexant was approved in the United States under the trade name Belsomra for the treatment of insomnia. During regulatory review, the Food and Drug Administration expressed concerns about potential next-day residual effects (Citrome, 2014; Vermeeren et al., 2015) associated with higher Belsomra doses as a consequence of its extended plasma half-life. This episode highlights the importance of identifying compounds with an optimal pharmacokinetic and pharmacodynamic (PK/PD) profile in man during the discovery process of an insomnia drug.

From a drug discovery perspective, the identification of candidate molecules for the treatment of insomnia poses particular challenges. Beyond pharmacological target potency and brain penetration as basic requirements, rapid onset and adequate sleep duration are key features. For compounds with little interspecies differences in receptor pharmacology and pharmacokinetics, appropriate drug candidates might be readily identified on the basis of animal pharmacology data. Translational aspects become more important for molecules exhibiting pronounced interspecies differences in drug disposition. For example, plasma protein binding affects drug distribution into tissues and total blood clearance, and thus influences the pharmacokinetic half-life of a drug. Physiology-based pharmacokinetic and pharmacodynamic (PBPK-PD) modeling has been established as an appropriate tool to translate animal PK and PD data into man early in drug development (Jones et al., 2009). The present report describes the central and early role of PBPK-PD modeling in drug discovery as a key tool in the selection process of ACT-541468, a DORA that is currently in phase 2 clinical development for the treatment of insomnia disorders. Although this report focuses on only three prototypical compounds, the approach was in fact applied to a set of about 20 structural analogs from the same chemical class.

Materials and Methods

Chemicals and Reagents.

(S)-(2-(5-chloro-4-methyl-1H-benzo[d]imidazol-2-yl)-2-methylpyrrolidin-1-yl)(5-methoxy-2-(2H-1,2,3-triazol-2-yl)phenyl)methanone (ACT-541468), (S)-(2-(5-chloro-1,4-dimethyl-1H-benzo[d]imidazol-2-yl)-2-methylpyrrolidin-1-yl)(5-methyl-2-(2H-1,2,3-triazol-2-yl) phenyl)methanone (ACT-605143), (S)-(2-(6-bromo-4-methyl-1H-benzo[d]imidazol-2-yl)-2-methylpyrrolidin-1-yl)(5-methyl-2-(pyrimidin-2-yl)phenyl)methanone (ACT-658090) (Fig. 1), almorexant, suvorexant, and their corresponding salts were synthesized by the medicinal chemistry department of Actelion Pharmaceuticals Ltd (Allschwil, Switzerland) as described in Supplemental Fig. S1, or according to published methods (Baxter et al., 2011; Mangion et al., 2012; Boss et al., 2013; Verzijl et al., 2013; Boss et al., 2015a,b). Chemical purity of all compounds was in excess of 97%.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Chemical structures of ACT-541468, ACT-605143, and ACT-658090.

Orexin Receptor Antagonist Assays.

OxA curve shift experiments were performed to determine the surmountability of antagonism and to calculate the antagonistic potency [apparent inhibition constant as determined by a functional assay (Kb)] of the three benzimidazole compounds. Chinese hamster ovary (CHO) cells expressing the human or rat OX1 or OX2 receptors were grown in culture medium (Ham’s F-12 with l-glutamine; Life Technologies/Thermo Fisher Scientific, Waltham, MA) containing 300 μg/ml (for human receptor-expressing cells) or 1000 μg/ml (for rat receptor-expressing cells) G418, 100 IU/ml penicillin, 100 μg/ml streptomycin, and 10% heat-inactivated fetal calf serum (FCS). Prior to the experiment, the cells were seeded at 20,000 cells/well into 384-well black clear-bottom sterile plates (Greiner/Sigma-Aldrich, St. Louis, MO) and incubated overnight at 37°C in 5% CO2. On the day of the assay, culture medium in each well was replaced with 50 μl of staining buffer [1× Hanks’ balanced salt solution (HBSS), 1% FCS, 20 mM HEPES, 0.375 g/l NaHCO3, 5 mM of the organic anion transporter (OAT) inhibitor probenecid (Sigma-Aldrich), 3 μM of the fluorogenic calcium indicator fluo-4 AM (Life Technologies/Thermo Fisher Scientific), and 10% pluronic F-127 (Life Technologies/Thermo Fisher Scientific) ]. The cell plates were incubated for 1 hour at 37°C in 5% CO2 followed by equilibration at room temperature for ≥30 minutes. Fluo-4-stained cells were supplemented with 10 μl of 1:10 serially diluted 6-fold concentrated antagonist solution in the assay buffer (1× HBSS buffer, 0.1% bovine serum albumin, 20 mM HEPES, 0.375 g/l NaHCO3, pH 7.4) containing 0.6% dimethyl sulfoxide (DMSO). After 120 minutes, cells were stimulated with 10 μl of 1:5 serially diluted 7× OxA solution in assay buffer containing ≤0.1% methanol, resulting in final concentrations between 0.01 and 1000 nM. Calcium mobilization (proportional to OX receptor activation by OxA) was measured using the fluorescence imaging plate reader (FLIPR) assay (Tetra, excitation: 470–495 nm; emission: 515–575 nm; Molecular Devices, Sunnyvale, CA). To calculate the apparent Kb values, FLIPR traces were subjected to spatial uniformity correction and normalized by trace alignment at the last time point before agonist addition (ScreenWorks software; Molecular Devices). The maximum fluorescence signals per well were used to generate OxA concentration-response curves (CRCs) in GraphPad Prism (GraphPad Software, LaJolla, CA). The EC50 values and Hill slopes (n) for the OxA CRCs, and the IC50 values of the antagonists at approximately EC70 of OxA (1.6 nM for human OX1 and OX2, 8 nM for rat OX1 and OX2) were calculated using the proprietary IC50 Witch software (curve-intrinsic minima and maxima were used). The apparent Kb values were calculated via the generalized Cheng-Prusoff equation, using the on-day OxA EC50 value (EC50OxA) and the OxA concentration used for stimulation ([OxAstim]), with the following formula (Cheng and Prusoff, 1973; Miller et al., 1999):Embedded Image(1)Approximate receptor occupancy half-lives (ROt1/2) of the antagonists were determined using calcium release assays after antagonist washout. To this end, fluo-4-stained CHO cells were supplemented with 10 μl of 6× concentrated antagonists (serially diluted 1:10 in assay buffer with 0.6% DMSO). After 120 minutes, cells were washed extensively with assay buffer and then stimulated at several time points (0–63 minutes) with 10 μl of a 7× OxA solution, giving a final OxA concentration of EC70. Compound incubations and washout phase were performed at 37°C. Calcium mobilization was measured, and IC50 values were calculated and converted to the apparent Kb values via the generalized Cheng-Prusoff equation, using the OxA CRC slope determined at every time point, as described above (software settings: curve-intrinsic maximum; fixed minimum: full block by 10 μM suvorexant). The geometric means of Kb values generated in three independent experiments were then plotted on a semilogarithmic scale against time. The ROt1/2 was calculated from the Kb values obtained at 0 and 33 minutes after washout, assuming first-order dissociation kinetics, with the following formula:

Embedded Image(2)

Animal Housing.

Male, adult Wistar rats were used for pharmacokinetic [Crl:WI(Han) (Charles River, Sulzfeld, Germany)] and pharmacology experiments [RccHan:WIST (Harlan, Horst, The Netherlands) or Crl:WI(Han)]. All rats were maintained under standard laboratory conditions [temperature 20±2°C, relative humidity 55–70%, standard Provimi Kliba diet 3336 (Kaiseraugst, Switzerland, www.kliba-nafag.ch), and domestic mains tap water ad libitum] on a regular 12-hour light–dark cycle (lights on 6:00 AM). After arrival, rats were allowed at least one week of acclimatization to Actelion’s animal facility and were carefully monitored to ensure good health and suitability for inclusion in the study. Unless noted otherwise, rats were socially housed in groups of four in standard plastic rodent cages, and all tests were conducted during the light phase (8:00 AM to 6:00 PM) under illumination of >600 lx.

Pharmacokinetic experiments in the Beagle dog were performed in the Shanghai animal facilities of Actelion Pharmaceuticals Ltd in accordance with the Swiss animal protection law. The dogs were group-housed during the study and only separated for feeding and monitoring of clinical signs. With the exception of a period of fasting from the night before dose administration until 4 hours after dose administration, animals were given a daily allowance of 350 g of a standard laboratory diet and had free access to tap water.

All animals were housed in accordance with the National Institutes of Health guidelines and experimental procedures were approved by the Basel-Landschaft Veterinary Office and strictly adhered to Swiss federal regulations on animal experimentation. Well-being of animals was monitored during the day by trained technical staff. Animals were checked for body weight loss, abnormal breathing, pilo-erection, grooming, and locomotion as criteria of distress according to the company’s animal welfare policy and guideline on humane endpoints.

Pharmacokinetic Experiments in Rat and Dog.

Male Wistar rats (n = 3) with a body weight of ca. 200–250 g were used for pharmacokinetic experiments under license no. 169. For intravenous sampling, a jugular vein catheter was implanted 2 days prior to drug dosing under aseptic conditions. After recovery from general isoflurane anesthesia, animals were housed individually with free access to water and food during the recovery period and the entire duration of the experiment. Compounds for intravenous use were formulated as aqueous solution in 30% 2-hydroxypropyl-beta-cyclodextrin (HPβCD) starting from a 5% stock solution in DMSO. For oral gavage dosing, compounds were formulated as either a suspension in 0.5% methyl cellulose or as solution in PEG400.

Male Beagle dogs (n = 3) with body weights of 14.3–15.6 kg at the start of treatment were used in a crossover design with a washout period of 7 days under license number 2016-06-QCB-32. All experiments were performed in fasted state, and gastric pH was controlled by giving intramuscular pentagastrin at a dose of 6 μg/kg 20 minutes before and 30 minutes after oral dosing. For intravenous dosing, compounds were formulated as aqueous solution in 30% HPβCD starting from a 5% stock solution in DMSO. For the oral route, compounds were either suspended in 0.5% methyl cellulose, dissolved in PEG400, or given as capsules containing either mannitol or a 4:1 mixture of Cremophor RH40 and lauroglycol.

Serial blood samples of 0.25 ml (rats) or 2 ml (dogs) were taken over a period of 24 hours and transferred into vials fortified with EDTA as anticoagulant. Blood samples after oral dosing to rats were taken under light isoflurane anesthesia. Plasma was generated by centrifugation and stored at –20°C pending analysis.

Pharmacokinetic parameters were estimated using noncompartmental analysis within the Phoenix software package (version 6.4; Pharsight Corporation, Cary, NC). Area under the plasma concentration versus time curve extrapolated to infinity (AUC0–inf) was accepted if the percentage of area extrapolated to infinity (AUC%extp) did not exceed 20%, otherwise area under the plasma concentration versus time curve up to the last measurable concentration (AUC0–last) was reported.

Analytical Methods.

Samples from in vitro and in vivo experiments were fortified with 3–6 volume equivalents of methanol containing a close structural analog as internal standard, and proteins were removed by centrifugation at 3220 g for 20 minutes at 4°C. If required, supernatants were diluted with 1% aqueous formic acid or a 1:1 mixture of acetonitrile and 1% aqueous formic acid prior to quantification by liquid chromatography coupled to mass spectrometry (LC-MS/MS) on API5000 or API5500 mass spectrometers (AB SCIEX, Concord, Ontario, Canada) in selected reaction monitoring mode. Chromatographic separation was achieved on Phenomenex Luna C8, C18, Gemini C18 or Synergy Polar RP columns (4–5 μm, 2.0 × 20 mm internal diameter) operated at room temperature with a flow rate of 0.6 ml/min, using a linear gradient starting from 5 to 10% mobile phase B and a run time of 0.95 minutes. Mobile phases were 0.1% aqueous formic acid or 5 mM ammonium formate (pH 9, phase A) and acetonitrile or methanol (phase B).

Population—Based Pharmacokinetic Modeling.

Simcyp Population-based ADME Simulator (version 15; Sheffield, UK) was used for PBPK modeling, and simulation PBPK models were built on the basis of physicochemical, binding, permeability and metabolic stability data (Table 7; Supplementary Materials for experimental details). Oral absorption was modeled using the advanced dissolution, absorption, and metabolism (ADAM) model within Simcyp. Compounds were modeled as a solid formulation in immediate-release capsules for dog and human or as suspension for rat, using the diffusion-layer model of Wang and Flanagan (Jamei et al., 2009) to calculate the rate of dissolution from fine particles with an assumed diameter of 1 μm. The effect of bile salts upon dissolution and solubility was considered, matching the measured solubility in fasted state (FaSSIF) and fed state (FeSSIF) simulated intestinal fluids. Default values for particle density of 1.2 g/ml, a supersaturation ratio of 10, a precipitation rate constant of 15 minutes, and a diffusion layer thickness of 30 μm were assumed. A full PBPK, perfusion-limited distribution model was applied using method 2 within Simcyp, with an assumed tissue-to-plasma partition coefficient (Kp) scalar of 0.5 (Rodgers and Rowland, 2007; de Kanter et al., 2016). Elimination was assumed to be driven by CYP3A4-mediated metabolism, on the basis of evidence for almorexant (Dingemanse et al., 2014), suvorexant (Cui et al., 2016), and ACT-541468 (A.T., unpublished observation) using the well-stirred liver and ADAM gut model with fraction unbound in the gut (fu,gut) assumed to be 0.01. A factor of 2 was used to correct for the underprediction of in vitro–to–in vivo total plasma clearance (CL). Thus, the model was populated with measured intrinsic clearance (CLint) values from rat, dog, or human liver microsomes, set to occur by hepatic and intestinal cyp3a (rat), cyp3a12 (dog), or CYP3A4 (human), multiplied by 2, and corrected for nonspecific assay binding [fraction unbound in liver microsomes (fu,mic), Table 7]. OX2 occupancy was estimated using predicted unbound brain concentration versus time profiles, mean unbound Kb on the OX2 receptor (Tables 1 and 7), and a sigmoid Emax model with an Emax of 100% and a Hill slope of 1.Embedded ImageFree distribution of unbound drug between plasma and brain was assumed, as supported by the physicochemical properties of all compounds and the absence of relevant P—glycoprotein and breast cancer resistance protein–mediated efflux (A.T., unpublished observation).

View this table:
  • View inline
  • View popup
TABLE 1 

Mean apparent Kb values for selected dual orexin receptor antagonists in calcium release assays determined in CHO cells expressing human OX1 and OX2 receptors (n ≥ 3)

Ten virtual trials of 10 subjects each were simulated in a male, healthy volunteer population with an age range of 18–45 years. Variation of all physiologic parameters was on the basis of the default variation in the healthy volunteer population database used in Simcyp.

Brain Partitioning in the Rat.

Brain and plasma concentrations were determined 3 hours following oral administration at 30 and 100 mg/kg to rats (n = 3–4 per group) under license no.KV-BL-426. All compounds were formulated as solutions in PEG400. Blood was collected from the vena cava caudalis into plastic tubes containing EDTA as anticoagulant and centrifuged to yield plasma. Brain was collected after cardiac perfusion of 10 ml NaCl 0.9%. Brain tissue was then homogenized with 1 volume equivalent of cold phosphate buffer (pH 7.4), and compounds were extracted with methanol. Compound concentrations in plasma and brain were determined using LC-MS/MS as described above.

Telemetric Sleep/Wake Cycle Evaluation in Rats.

Sleep/wake cycles were evaluated on the basis of electroencephalography/electromyography (EEG/EMG) and home-cage activity recorded in individually housed Wistar rats under free-moving conditions under license no.KV-BL-205. Rats (250–350 g, 6–8 weeks of age) were equipped with telemetric transmitters (TL11M2-F20-EET; Data Science International, St. Paul, MN) that allowed the noninvasive detection of EEG/EMG and activity via signal transmission to a receiver. The surgical transmitter implantation was performed under aseptic conditions. A preoperative analgesia (buprenorphine 0.015 mg/kg s.c.) was administered 30 minutes before anesthesia. The entire surgical implantation was performed under general anesthesia [isoflurane inhalation (1–5% vol.) initiated in an anesthetic chamber]. The rat was placed and secured in a stereotaxic apparatus. The body of the transmitter was placed subcutaneously along the dorsal flank of the rat with the leads routed subcutaneously to an incision accessing the cranium. For EEG recordings, two trepanations were placed in the skull, 2 mm from either side of the midline and 2 mm anterior to the lambda suture for placement of one differential pair of electrodes. Two other superficial trepanations were drilled for screws as support for cementing the electrodes. The EMG leads were inserted in either side of the muscles of the neck and sutured into place. After surgery, rats recovered from anesthesia in a chamber equipped with a heating pad at 37°C. They were then housed individually and received analgesia with 0.015 mg/kg s.c. of buprenorphine B.I.D. for the first 2 days of the 2-week postsurgery recovery period. Before the experiment, rats were acclimatized for 3 days in their home cages placed in ventilated sound-attenuating boxes, on a regular 12-hour light/dark cycle. Experiments used a “crossover” design, i.e., each animal was alternatively treated with a test compound and the vehicle, with at least 72 hours between administrations. Treatments were administered orally (gavage) at the transition between the day and the night phase. Each telemetric recording covered a 24-hour “baseline” period preceding the treatment, the 12-hour night period immediately following the administration of treatment, and a 36-hour recovery (washout) period. The recordings were split into a sequence of equal intervals of user-defined length (10 seconds). Sleep and wake stages (active wake, quiet wake, non-REM sleep, or REM sleep) were assigned to each interval automatically using the Somnologica Science software (Medcare; Embla Systems Ltd., Thornton, CO) on the basis of frequency estimation for EEG, amplitude discrimination for the EMG, and the locomotor activity, as follows. Wake was characterized by low-amplitude EEG activity with relatively greater power in the higher frequency bands, such as alpha-band (10–13 Hz), accompanied by moderate to high levels of EMG activity. Active and quiet wake were distinguished on the basis of the locomotor activity and the amplitude of the EMG. Non-REM sleep was defined by high-amplitude EEG activity with greater power in the delta frequency band (0.5–5 Hz) and low EMG activity. REM sleep was characterized by low-amplitude EEG activity focused in the theta frequency band (6–9 Hz) and no EMG activity. Data were analyzed by two-tailed paired Student’s t test or one-way analysis of variance (ANOVA) followed by the posthoc Tukey’s multiple comparisons test, using GraphPad Prism (GraphPad Software). Differences were considered statistically significant at P < 0.05.

PK and PD Assessments of ACT-541468 in Healthy Subjects.

The single-dose pharmacokinetic profile of ACT-541468 after daytime and night-time administration was assessed in the two single-center, double-blind, placebo-controlled, randomized studies AC-078-101 (NCT02919319) and AC-078-102 (NCT02571855). Both trials were conducted in full compliance with the principles of the Declaration of Helsinki, and study protocols were approved by independent Ethics Committees. Detailed information about study endpoints and inclusion and exclusion criteria are available at www.clinicaltrials.gov. For daytime pharmacokinetic assessments, eight healthy male subjects received either placebo (n = 2) or ACT-541468 (n = 6) at doses of 5, 25, 50, 100, and 200 mg, given in the morning after overnight fasting. Blood samples were collected over a period of 96 hours postdose, plasma was separated by centrifugation, and ACT-541468 concentrations therein determined by LC-MS/MS. CL and apparent volume of distribution at steady-state (Vss) were determined using an intravenous 14C-ACT-541468 microdose given on top of 100 mg oral ACT-541468 (n = 4). Plasma levels of radiolabeled ACT-541468 were quantified by accelerator mass spectrometry technique after chromatographic separation. Pharmacokinetic parameters after night-time administration were assessed in 6 healthy subjects who received either 25 mg ACT-541468 (n = 4) or placebo (n = 2). PD effects were assessed in study AC-078-101 at all doses after daytime administration of ACT-541468 during the first 10 hours after dosing, using a battery of objective tests including saccadic eye movements, adaptive tracking, and body sway.

Results

Inhibition of Orexin Receptor Signaling in Cellular Assays

Almorexant and suvorexant have shown efficacy in inducing sleep in animals and humans, and, therefore, served as benchmark for the present benzimidazole series. Table 1 and Fig. 2 summarize the characterization of the three benzimidazoles ACT-541468, ACT-605143, and ACT-658090 in calcium-release assays in comparison with almorexant and suvorexant. The IC50 values measured at approximately EC70 of OxA (1.6 nM) were used to calculate the apparent Kb values. All five compounds behaved as potent dual antagonists, and displayed an insurmountable profile at both receptors. Kb values ranged from 0.52 to 8.3 nM at human OX1 receptors, and from 0.42 to 5.9 nM at human OX2 receptors. Kb values on the corresponding rat and human receptors were comparable (Supplemental Table S1). Using calcium-release assays after antagonist washout, ROt1/2 on OX1 was estimated to be 6–9 minutes for suvorexant and all benzimidazoles, and 22 minutes for almorexant (Table 2). On OX2, suvorexant, ACT-541468 and ACT-605143 showed ROt1/2 in the range of 4–6 minutes, whereas ROt1/2 was 13–14 minutes for ACT-658090 and almorexant. In summary, the three benzimidazoles displayed in vitro receptor interaction profiles similar to those of suvorexant and almorexant. ACT-541468 was a selective OX1 and OX2 receptor antagonist in a panel screen of more than 130 established central and peripheral pharmacological targets (A.T., unpublished observation).

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Effect of dual orexin receptor antagonists on OxA-induced calcium flux in CHO cells expressing recombinant human OX1 or OX2 receptors. Cells were stained with fluo-4 and preincubated with dilution series of antagonists for 120 minutes followed by the addition of a dilution series of OxA. Peak fluorescence values were converted to CRCs. Data shown are the average of duplicate values (± S.D.) from a representative of at least three experiments. IC50 values at 1.6 nM OxA were determined and used as a basis to calculate the apparent Kb via the generalized Cheng-Prusoff equation.

View this table:
  • View inline
  • View popup
TABLE 2 

Receptor occupancy half-lives of orexin receptor antagonists at human OX1 and OX2 receptors as determined by calcium flux measurements after compound washout

Pharmacology in the Rat

Brain Penetration.

Brain penetration of ACT-541468, ACT-658090, and ACT-605143 was determined 3 hours after administration of oral doses of 30 mg/kg and 100 mg/kg. At 30 mg/kg, mean total brain concentrations of ACT-541468 and ACT-658090 reached 665 and 614 nM but was only 154 nM for ACT-605143 (Table 3). Total brain levels increased in a more than dose-proportional manner at 100 mg/kg, reaching 2247–12,000 nM. Correction for in vitro brain binding (fu,brain; Table 7) yielded unbound brain concentrations around or above the Kb values on rat OX2 (Supplemental Table S1), warranting further investigations in a rat sleep model.

View this table:
  • View inline
  • View popup
TABLE 3 

Brain partitioning of orexin receptor antagonists in Wistar rats after oral dosinga

Effect on Sleep/Wake Cycles in Rats.

All three benzimidazole DORAs were tested in male Wistar rats implanted with telemetric transmitters to allow for continuous EEG/EMG-based evaluation of sleep/wake cycles in freely moving animals in their home cages. ACT-541468, ACT-605143, and ACT-658090 were dosed at 30 mg/kg at the beginning of the night-active phase, when endogenous orexin levels are rising.

All three compounds showed robust effects on sleep/wake stages over the 6-hour period postdose (Fig. 3). ACT-541468 decreased the time spent in active wake by 22% compared with vehicle-treated rats (−45 minutes; P = 0.0068, paired t test) and increased the time spent in non-REM and REM sleep by 29 and 84% (+26 and +10 minutes; P = 0.0009 and P = 0.0228), respectively. Latency to persistent non-REM sleep significantly decreased by 59% from 44 to 18 minutes (P = 0.0002), and to REM sleep by 58% from 71 to 30 minutes (P = 0.0006, Fig. 4). A similar effect was observed with ACT-605143, which decreased the time spent in active wake by 21% compared with vehicle-treated rats (−45 minutes; P = 0.0089), and increased the time spent in non-REM and REM sleep by 34 and 146% (+24 and +16 minutes; P = 0.0092 and P < 0.001), respectively. Latency to persistent non-REM sleep significantly decreased by 49% from 35 to 18 minutes (P = 0.0033, Fig. 4), and to REM sleep by 44% from 62 to 35 minutes (P = 0.007). Finally, ACT-658090 decreased the time spent in active wake by 25% (−46 minutes; P < 0.001) and increased the time spent in quiet wake by 21% (+10 minutes; P = 0.0099). It increased the time spent in both non-REM and REM sleep by 27% and 57% (+26 minutes, P = 0.0001; and +10 minutes, P = 0.0127), respectively. Latency to persistent non-REM sleep significantly decreased by 50% from 42 to 21 minutes (P = 0.030), and to REM sleep by 54% from 84 to 39 minutes (P = 0.032). Efficacy of the 3 benzimidazole DORAs over the 6-hour period following a 30 mg/kg dose was similar (Fig. 4). The increase in total sleep time over the 6-hour period was between 36 and 40 minutes for all compounds and differences between compounds were not statistically significant (P = 0.9087, one-way ANOVA). Drug exposure, expressed as AUC0–last, was also similar, within a 2.5-fold range from 2950–7510 nM⋅h (Table 4).

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Effect of benzimidazole DORAs on sleep/wake stages at 30 mg/kg in telemetrized rats. Effects of ACT-541468, ACT-658090, and ACT-605143 during the first 6 hours of the night-active period in telemetrized male Wistar rats (% of total time). Data are expressed as mean ± S.E.M., n = 8 per group. *P < 0.05, **P < 0.01, ***P < 0.001 compared with matched vehicle-treated rats.

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Effect of benzimidazole DORAs on total sleep time (A), latency to non-REM (B), and latency to REM sleep (C) at 30 mg/kg in telemetrized rats. Effects of ACT-541468, ACT-658090, and ACT-605143 on (A) total sleep time during the first 6 hours of the night-active period (difference versus vehicle in minutes for all parameters), (B) on latency to persistent non-REM sleep (in minutes, first episode of at least 60 seconds), and (C) latency to persistent REM sleep (in minutes, first episode of at least 30 seconds) in telemetrized male Wistar rats. Data are expressed as mean ± SEM, n = 8 per group, *P < 0.05, **P < 0.01, ***P < 0.001 compared with matched vehicle-treated rats.

View this table:
  • View inline
  • View popup
TABLE 4 

Observed and simulated mean pharmacokinetic parameters of orexin receptor antagonists after oral dosing to Wistar rats and Beagle dogsa

The dose response of ACT-541468 in the rat sleep model was characterized at 10, 30 and 100 mg/kg (Fig. 5). ACT-541468 dose-dependently impacted sleep/wake parameters. Over the 6-hour time period following administration, time spent in active wake decreased by 6–50 minutes, and time spent in non-REM and REM sleep increased by 2–29 minutes and 5–15 minutes, respectively. The lowest dose giving significant effects was 30 mg/kg. In the vehicle group, latency to persistent non-REM sleep varied from 40 to 47 minutes, depending on the group. Compared with vehicle, ACT-541468 significantly decreased the latency to persistent non-REM sleep to 18 minutes at 30 (P = 0.0002) or 100 mg/kg (P = 0.0079), and the latency to persistent REM sleep down to 30 minutes and 37 minutes at 30 mg/kg (P = 0.0006) and 100 mg/kg (P = 0.014), respectively.

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Dose-response of single oral administrations of ACT-541468 on sleep/wake stages in telemetrized rats (A), latency to persistent non-REM (B) and REM sleep (C) at 10, 30, and 100 mg/kg. Effect of ACT-541468 on time spent in active wake, quiet wake, non-REM, and REM sleep during the first 6 hours of the night-active period (A), on latency to persistent non-REM sleep (in minutes, first episode of at least 60 seconds, B), and on latency to persistent REM sleep (in minutes, first episode of at least 30 seconds, C) in telemetrized male Wistar rats (in minutes, difference versus matched vehicle). Data are expressed as mean ± S.E.M., n = 8 per group. *P < 0.05, **P < 0.01, ***P < 0.001 compared with matched vehicle-treated rats.

In the vehicle-treated animals, the proportion of non-REM and REM sleep relative to total sleep time varied between 79–86% for non-REM sleep, and between 14–21% for REM sleep (Table 5). The proportion of non-REM and REM sleep did not change in a statistically significant manner for ACT-541468 at 30 mg/kg (P = 0.15) or 100 mg/kg (P = 0.59), or for ACT-658090 at 30 mg/kg (P = 0.27). In contrast, the time spent in non-REM sleep significantly decreased from 86 to 78% (P = 0.00074) for ACT-605143 at 30 mg/kg.

View this table:
  • View inline
  • View popup
TABLE 5 

Proportion of non-REM and REM sleep relative to total sleep time over the first 6-hour period following oral dosing of ACT-541468, ACT-605143, or ACT-658090

Pharmacokinetics in Rat and Dog

Pharmacokinetics of all three benzimidazole compounds were characterized in male rats and dogs to generate a database for validation of the respective PBPK models. Pharmacokinetic results after oral and intravenous administration are summarized in Tables 4 and 6, respectively. Plasma clearance of ACT-605143 and ACT-658090 in the rat was 32–36 ml/min per kilogram, and 85 ml/min per kilogram for ACT-541468. After correction for blood/plasma partitioning (Table 7), blood clearance of all three compounds was in a narrow range of 36–45 ml/min per kilogram, i.e., ca. 51–64% of liver blood flow in the rat. Plasma and blood clearances in the dog were 3.9–9.5 ml/min per kilogram and 6.5–15 ml/min per kilogram, respectively, the latter corresponding to 44–48% of liver blood flow for ACT-541468 and ACT-605143 but only 21% for ACT-658090. Vss in the rat was in large excess of total body water, indicating significant drug distribution into tissues: 3.2 and 3.9 l per kilogram for ACT-541468 and ACT-658090, but only 0.8 l/kg for ACT-605143. Vss in the dog was in a more narrow range, between 1.1–2.4 l/kg.

View this table:
  • View inline
  • View popup
TABLE 6 

Observed and simulated mean systemic pharmacokinetic parameters of orexin antagonists in rats, dogs, and healthy subjectsa

View this table:
  • View inline
  • View popup
TABLE 7 

Biochemical and physicochemical data for PBPK model development

Biochemical data are means of at least triplicate experiments.

Oral absorption of all compounds was rapid in rat and dog, with peak plasma concentrations (Cmax) being reached within 0.25–2.3 hours (Table 4). Oral exposures (AUC0–last) in the rat at 10 mg/kg varied by about 10-fold, from 412 nM⋅h for ACT-541468 to 4060 nM⋅h for ACT-658090, which was also reflected in their different oral bioavailability (F) of 9 versus 43%. Smaller differences in AUC0–last were observed in the dog, with 2020 nM⋅h for ACT-541468 and 5610 nM⋅h for ACT-658090. Similar trends were evident for Cmax in both species (Table 4).

PBPK Model Development for Predicting Human PK Profiles

PBPK models for the benzimidazole DORAs ACT-541468, ACT-605143, and ACT-658090 were built on the basis of in silico and biochemical data (Table 7), as outlined in Fig. 6. PBPK models were initially developed for rat and dog and validated against observed PK data in both species. All three PBPK models consistently overpredicted observed Vss. Therefore, a Kp scalar of 0.5 was applied to empirically correct the predicted tissue-to-plasma ratios in all simulated tissues. In contrast, CL predictions underestimated observed values by approximately 2-fold, and microsomal CLint was therefore multiplied by 2 in the PBPK models. The simulated pharmacokinetic parameters of these refined PBPK models after intravenous and oral dosing in rat and dog were considered similar enough to the observed values to construct the corresponding human PBPK models (Table 4 and Table 6). The simulated plasma concentration versus time profiles and derived pharmacokinetic parameters of the three benzimidazoles in healthy volunteers at a dose of 25 mg are shown in Fig. 7A. At this dose, the predicted fraction absorbed was 96–100% and not sensitive to the supersaturation ratio (1–100) or precipitation constant (0.4–40). Among the three compounds, ACT-541468 showed the fastest simulated oral absorption with a time to reach peak plasma concentration (Tmax) of 2 hours, the shortest initial half-life of 4.3 hours, and the lowest Cmax and AUC0–24h of 814 nM and 6500 nM⋅h, respectively (Table 8). ACT-658090 and ACT-605143 exhibited 5.9- to 7.3-fold higher AUC0–24h, and 2.9–3.3 times higher Cmax, and reached peak plasma concentrations later than ACT-541468, with Tmax of 3.6–3.9 hours. Most important for a sleep agent, plasma levels of ACT-658090 and ACT-605143 declined with relatively long half-lives of 10.4 and 26.8 hours, resulting in significant residual drug levels at the end of the 24-hour dosing interval (Fig. 7A).

Fig. 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 6.

PBPK and absorption model development and application.

Fig. 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 7.

Simulated geometric mean plasma concentration (A) and OX2 receptor occupancy (B) versus time profiles of ACT-541468, ACT-605143 and ACT-658090 after a 25-mg dose given to healthy male subjects. Plasma concentration-versus-time profiles were predicted using PBPK models (A), and OX2 receptor occupancy-versus-time profiles (B) were predicted on the basis of a Emax model driven by the unbound brain concentrations of the benzimidazole compounds.

View this table:
  • View inline
  • View popup
TABLE 8 

Observed and simulated pharmacokinetic parameters and estimated OX2 receptor occupancies in healthy male subjectsa

PBPK models for suvorexant and almorexant were developed using physicochemical and biochemical data (Supplemental Table S2) together with published clinical data for both compounds (Hoch et al., 2012; Sun et al., 2013; Dingemanse et al., 2014; TGA, 2014). For both, suvorexant and almorexant, oral absorption was modeled using ADAM and cellular permeability determined in MDCKII cells. Tissue distribution of suvorexant and almorexant was modeled as described above, and elimination was modeled on the basis of published plasma clearances of 5 l/h and 43 l/h, respectively (Hoch et al., 2012; TGA, 2014). Final model parameters after intravenous and oral dosing are presented in Tables 6 (CL and Vss) and 8 [AUC0–24h, Cmax, Tmax, initial half-life (T1/2)]. The observed and simulated mean plasma concentration–time profiles of 40/50 mg suvorexant and 200 mg almorexant are shown in Fig. 8.

Fig. 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 8.

Observed (daytime and night-time) and simulated geometric mean plasma concentration-versus-time plots of 200 mg almorexant (A) and 40 or 50 mg suvorexant (B). Plasma concentration versus time profiles were reconstructed on the basis of published data for almorexant (Hoever et al., 2012a) and suvorexant (Sun et al., 2013, 2015). Horizontal lines indicate total plasma concentrations at 8 hours after night-time administration. Data for almorexant are given as geometric mean ± S.D. with daytime time points being shifted by +10 minutes for clarity. Data for suvorexant are given as geometric means only as no information on variability is given in the published literature.

Human Dose Estimates

PK/PD Analysis of Almorexant and Suvorexant in Rats as Basis for Human PD Threshold Concentrations.

An attempt to estimate drug exposures needed to induce sleep in humans was done on the basis of a PK/PD analysis in rats. The sleep-wake pattern in rats was analyzed after treatment with almorexant and suvorexant at doses of 10, 30, and 100 mg/kg given at the beginning of the active period (lights off), together with exposure data from PK experiments in a satellite group of rats. Both compounds were inactive at 10 mg/kg (Supplemental Fig. S2), with almorexant and suvorexant plasma exposures in the first hours reaching 97 and 443 nM, respectively. At 30 mg/kg, total sleep time increased significantly with both compounds to about 3–7 hours for almorexant, and about 2 hours for suvorexant (Supplemental Figs. S2 and S3) at total and unbound plasma concentrations of ca. 195 and 1.2 nM for almorexant and ca. 890 and 3.5 nM for suvorexant, which were regarded as PD threshold plasma concentrations in rat. Considering human plasma protein binding and the absence of potency differences at target receptors (Table 1 and Supplemental Table S1), and assuming the need for similar degrees of OX2 blockade in rats and humans, the predicted human PD threshold plasma concentrations would be 195 nM for almorexant and 3550 nM for suvorexant. These projected values were about 5- to 6-fold above the exposures observed in man with sleep-inducing doses of almorexant (200 mg; 8-hour residual exposure of 20 ng/ml) and suvorexant (50 mg; 8-hour residual exposure of 240 ng/ml; Fig. 8). It is noteworthy, that an exact determination of sleep duration in rats is technically difficult and is a methodological limitation of this approach. Moreover, the fragmented sleep pattern in rats, i.e., alternating sleep and wake periods both in the active night and the inactive day period might render a direct translation of PD threshold concentrations from rat to humans difficult. Owing to these uncertainties, human dose predictions were performed on the basis of OX2 receptor occupancy estimates.

Orexin 2 Receptor Occupancy Estimates of Almorexant and Suvorexant in Humans.

On the basis of PBPK models of almorexant and suvorexant (Table 8 and Fig. 8) and assuming OX2 blockade as the driving force for sleep maintenance, PBPK-PD models, i.e., OX2 receptor occupancy versus-time profiles, were constructed using a sigmoid Emax model, unbound drug concentrations in brain, and the unbound Kb values for OX2 inhibition shown in Table 1. OX2 receptor occupancies were then estimated at 8 hours postdose as the presumed end of the sleep period (Table 8). According to these PBPK-PD models, 200 mg almorexant resulted in a peak OX2 receptor occupancy of 81% at 2.5 hours postdose, which declined to 63% at 8 hours postdose. Likewise, a 50-mg dose of suvorexant yielded a maximal OX2 receptor occupancy of 73% reached at 3.2 hours after drug intake, which slowly declined to 66% at 8 hours postdose.

Human Dose Estimates for Benzimidazole DORAs.

The OX2 receptor occupancy versus time profiles of the three benzimidazole DORAs in healthy male subjects were derived starting from the respective PBPK models (Fig. 7A). OX2 receptor occupancy versus time profiles in brain (Fig. 7B) were then predicted on the basis of unbound brain concentrations, assuming that unbound drug in plasma readily equilibrates with brain and using the unbound Kb constants on OX2 in a sigmoid Emax model with a Hill factor of 1. Using this approach, ACT-541468 doses were identified targeting an OX2 receptor blockade of about 65% over a period of 8 hours. As shown in Table 8, an ACT-541468 dose of 25 mg resulted in a predicted OX2 receptor occupancy of 58% at 8 hours postdose. OX2 receptor occupancies estimated for ACT-605143 and ACT-658090 at 8 hours postdose at the same 25 mg dose yielded values of 53 and 14%, respectively (Table 8).

ACT-541468 Pharmacokinetics and Pharmacodynamics in Healthy Subjects.

The pharmacokinetic and pharmacodynamic profile of ACT-541468 was assessed in healthy subjects (n = 6 on active, n = 2 on placebo) after daytime administration at doses ranging from 5 to 200 mg. Night-time pharmacokinetic data were only generated for the 25 mg dose (n = 4). Only data on the 25 mg dose are reported here. Geometric mean pharmacokinetic parameters (median for Tmax) after daytime and night-time administration are summarized in Table 8 and are graphically depicted in Fig. 9. After daytime administration, ACT-541468 was rapidly absorbed as judged from the median Tmax of 1 hour. Peak plasma concentrations and AUC0–24h were 1400 nM and 5700 nM⋅h, respectively. Oral absorption was slightly delayed to 1.5 hours after night-time dosing and peak plasma concentrations were reduced to 1050 nM. AUC0–24h was 8270 nM⋅h, i.e., about 45% higher compared with daytime administration. The pharmacodynamic effects of ACT-541468 were assessed during the first 10 hours after dosing using a battery of objective tests, including saccadic eye movements, adaptive tracking, and body sway. The effect of 25 mg ACT-541468 on adaptive tracking as one of the most sensitive assessments of vigilance and attention is depicted in Fig. 10.

Fig. 9.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 9.

Observed (daytime and night-time) and simulated geometric mean plasma concentration-versus-time profiles of ACT-541468 after a 25 mg dose administered to healthy subjects. Plasma concentration versus-time profiles were either predicted using PBPK model (simulated) or observed in a clinical trial (daytime and night-time) (Muehlan et al., 2017). Observed data are given as geometric means ± S.D., with daytime time points being shifted by +10 minutes for clarity.

Fig. 10.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 10.

Adaptive tracking performance of healthy male subjects after a 25 mg dose of ACT-541468. Effect of 25 mg ACT-541468 in an adaptive tracking test in healthy male subjects (n = 6) over a period of 10 hours postdosing, expressed as mean change ± S.D. from baseline, and compared with placebo (n = 2).

CL and Vss were determined from an intravenous 14C-ACT-541468 microdose given on top of 100 mg oral ACT-541468 (n = 4), followed by quantification of ACT-541468 using accelerator mass spectrometry technique after chromatographic separation. Results are summarized in Table 6 and were used for the validation of the human PBPK model.

Discussion

An ideal sleep drug should mediate rapid sleep onset and sleep maintenance through major parts of the night and be devoid of next-day effects. Although sleep onset may be optimized by appropriate formulation strategies, including particle-size control or use of solubility enhancers, sleep maintenance is dependent on intrinsic factors such as the time course of target receptor blockade in the brain. Tools allowing for robust estimates of drug levels in human brain are therefore key for a successful sleep drug discovery program. Sleep models in rodent and nonrodent animal species have been conventionally used for this purpose and constitute an appropriate choice for compounds with little interspecies differences in receptor pharmacology and pharmacokinetics. Translation of animal data to man becomes challenging when interspecies differences are prominent.

All three benzimidazole DORAs described here fulfill the basic requirements of a sleep drug. They are potent and insurmountable antagonists of both orexin receptors showing comparable Kb values on OX1 and OX2 (Table 1). In line with their physicochemical properties and the absence of relevant P—glycoprotein efflux, they readily cross the blood-brain barrier, as evidenced by the similarity of unbound brain and plasma concentrations in rats (Table 3). A 30 mg/kg dose increased non-REM, REM, and total sleep time in telemetrized rats and decreased the latency to the first episode of persistent non-REM and REM sleep compared with vehicle-treated animals. The pharmacological profiles in the rat were similar to each other and comparable to other DORAs described in literature (Aissaoui et al., 2008; Boss et al., 2008, 2014; Cox et al., 2010; Sifferlen et al., 2010, 2013, 2015; Winrow et al., 2012; Heidmann et al., 2016). No clear differentiation between compounds was therefore possible on the basis of drug efficacy in rats.

In contrast, the benzimidazoles exhibited significant interspecies differences in key parameters driving drug distribution and clearance (Table 7). Metabolic stability in liver microsomes differed by 7.2- to 14-fold between rat and human, whereas plasma protein binding, the major determinant for general tissue and specifically brain penetration, differed by 9- to 30-fold. Ratios of unbound brain and plasma concentrations in the rat were below unity and increased with dose (Table 3). Nonlinear plasma protein binding was demonstrated for ACT-541468 at concentrations relevant for rat pharmacology assessments but not clinical testing (Supplemental Table S3), which might, at least in part, contribute to the dose-dependent brain/plasma ratios. Other factors, such as P-gp or breast cancer resistance protein–mediated efflux might also play a role in rats but have been excluded for man. Pharmacokinetic behavior was therefore expected to significantly differ between rat and man, calling for a tailored approach for the identification of a candidate drug exhibiting, beyond rapid sleep onset and efficacy, a sleep maintenance in man not longer than 8 hours.

The pharmacokinetic differences predicted for these benzimidazoles are mostly a reflection of differences in drug CL and tissue distribution (Table 6 and Fig. 7A). ACT-541468 had a 6–12 times higher simulated CL compared with ACT-605143 and ACT-658090 (Table 6), resulting from its CLint in liver microsomes and the lowest binding to plasma and liver microsomal proteins (Table 7). On the other hand, ACT-541468 exhibited a larger simulated volume of distribution, i.e., 0.5 l/kg versus 0.2 l/kg, in line with its lower plasma protein binding and distribution coefficient (logD). The combination of higher clearance and larger volume of distribution resulted in a simulated initial ACT-541468 plasma half-life of only 4.3 hours, which favorably compared with the 10.4 hours and 26.8 hours of ACT-605143 or ACT-658090, respectively, and also to suvorexant with a simulated half-life of 8.7 hours. It is noteworthy, that these differences in pharmacokinetic profiles were not obvious from simple inspection of the individual modeling input data such as CLint, plasma, or microsomal binding, pKa and logD, which were all within a 2- to threefold range.

Published data on human pharmacodynamics and pharmacokinetics of 200-mg almorexant (Hoever et al., 2012a) and 50-mg suvorexant (Sun et al., 2013) after night-time administration were used as a basis to estimate active doses of the benzimidazoles in man. Initial inspection of the plasma concentration versus time profiles of both compounds at the end of the presumed 8-hour sleep period indicated residual almorexant and suvorexant plasma concentrations of 20 ng/ml and 240 ng/ml, respectively (Fig. 8). This 12-fold difference in total plasma concentrations between both compounds appeared somewhat surprising in light of apparently identical OX2 receptor potency in cellular assays (Table 1). Introducing corrections for molecular weight, plasma protein binding, and non-specific binding in the orexin binding assays (Table 7) yielded comparable unbound almorexant and suvorexant concentrations of 1.5 and 2.3, when expressed as multiples of their respective unbound Kb values at OX2. PBPK-PD models constructed for both antagonists yielded consistent levels of OX2 receptor occupancy of 63–66% at the end of an 8-hour period (Table 8). A very similar threshold of 65–80% was recently reported for other DORAs in rat and dog (Gotter et al., 2013). Although almorexant and suvorexant both exhibited the same degree of OX2 receptor blockade at 8 hours postdose, only suvorexant was associated with impaired next-day performance (Citrome, 2014; Vermeeren et al., 2015). In addition to the absolute values in OX2 blockade, their rate of decline at the end of the sleep period was identified as a second key factor determining the absence of next day effects. The 65% OX2 threshold was then used as guidance for predicting active doses of the benzimidazoles (Fig. 7B). The time course of OX2 receptor blockade generally followed plasma concentrations for all three compounds. Major differences were, however, observed in the magnitude of OX2 blockade. Despite the lowest simulated total plasma exposure, ACT-541468 elicited the highest peak OX2 receptor blockade of 79%, whereas only 59% was reached with ACT-605143. Differences in plasma protein binding compensated for the 2.9-fold higher total plasma concentrations of ACT-605143, resulting in almost equal unbound peak concentrations of ACT-541468 in brain, i.e., 1.1 nM versus 0.96 nM. At 8 hours postdose, OX2 receptor occupancies were 58% for ACT-541468 and 53% for ACT-605143, indicating that both compounds met the requirement to maintain sleep over an 8-hour period. Slightly lower OX2 receptor occupancies than those deduced from clinical data on almorexant and suvorexant were targeted on purpose to compensate for the higher drug concentrations expected after night-time dosing (Hoever et al., 2012a; Sun et al., 2013), which PBPK-PD modeling was technically unable to capture. Compared with ACT-605143, the decline in OX2 receptor occupancy was faster with ACT-541468, in line with the higher CL, lower Vss, and resulting shorter half-life (Table 6). On the basis of PBPK-PD modeling, ACT-658090 was predicted to elicit a maximum OX2 receptor blockade of only 15%. This somewhat surprising outcome in light of the excellent pharmacokinetic profile (Fig. 7A) is best explained by the lower OX2 receptor potency combined with very high plasma binding (Table 7), resulting in insufficient unbound drug levels in brain to elicit relevant OX2 receptor blockade. On the basis of above analysis and its overall favorable profile in terms of animal pharmacology, early safety and drug-drug interaction potential (A.T., unpublished observation), ACT-541468 was selected for clinical development.

The pharmacokinetic profile of ACT-541468 in healthy subjects was characterized after daytime and night-time administration in two Phase 1 studies, whereas its pharmacodynamics were assessed during the first 10 hours after daytime administration using a battery of objective and subjective assessments. Observed PK parameters at a dose of 25 mg (Table 8 and Fig. 9) indicated rapid oral absorption after daytime administration, with peak plasma concentrations of 1400 nM being reached within 1 hour. Plasma AUC0–24h was well predicted within a 14% range. The observed initial half-life was 3.0 hours and thus slightly shorter than the simulated value of 4.3 hours, well in accordance with the higher observed CL, i.e., 1.0 ml/min per kilogram versus predicted 1.2 ml/min per kilogram, and lower Vss, 0.4 l/kg versus 0.5 l/kg (Table 6). The terminal half-life of ACT-541468 was about 6 hours (Muehlan et al., 2017). After night-time dosing of ACT-541468, mean peak plasma concentrations only reached 1050 nM, whereas AUC0–24h increased by 27% to 8270 nM⋅h. Oral absorption was slightly delayed, with a Tmax of 1.5 hours, and plasma half-life increased from 3.0 to 5.1 hours. The differences between ACT-541468 daytime and night-time pharmacokinetics may result from differences in physiology between night and day such as intestinal motility and perfusion, or liver blood flow. It is important to note, that the observed differences in ACT-541468 PK were rather small, in contrast to the significantly delayed oral absorption and reduced plasma levels after night-time administration of almorexant (Table 8 and Fig. 8) (Hoever et al., 2012a). From a methodological point of view, it is noteworthy that PK simulations were run with a total of 100 subjects, whereas the observed PK data are means of only six and four healthy subjects, which might not properly represent a larger population. Pharmacodynamically, 25 mg ACT-541468 revealed significantly reduced vigilance and attention (Fig. 10) and had clear effects on visuomotor coordination and postural stability. Onset of effects was observed within 1 hour, and returned to baseline within 3–6 hours. These early assessments in healthy subjects after daytime dosing suggest that a 25-mg dose has the potential to elicit the desired sleep effects in insomnia patients.

In conclusion, we have described the discovery of the dual orexin receptor antagonist ACT-541468 on the basis of early use of PBPK and PD modeling as part of the lead optimization process. ACT-541468 was identified out of a larger set of structurally related derivatives on the basis of its simulated OX2 receptor occupancy, targeting for an effect duration of about 8 hours at a 25 mg dose, and a sufficiently short half-life to minimize next-day effects. Phase I clinical trials with ACT-541468 in healthy subjects confirmed PBPK model-based predictions, as ACT-541468 reached peak plasma concentrations within 1 hour after dosing and exhibited an initial plasma half-life of only 3 hours. Drug exposure at a dose of 25 mg reduced vigilance and attention, and impaired visuomotor coordination and postural stability in healthy subjects after daytime administration. Differences between night-time and daytime PK performance may result from changes in physiology between day and night but are not expected to negatively impact on performance of ACT-541468 in insomnia patients.

Acknowledgments

The authors thank Stephane Delahaye, Susanne Globig, Bruno Capeleto, Charlyse Herrmann, Pascal Rebmann, Daniela Kruesi, Marc Candreia, Ursula Fusco-Hug, Thomas Sasse, Katalin Menyhart, Celia Müller, Cédric Fischer, Hélène Massinet, and Hélène Roellinger for their dedication and expert experimental contributions. The authors thank Alexey Veligodskiy for help in the writing of the manuscript.

Authorship Contributions

Participated in research design: Treiber, de Kanter, Roch, Gatfield, Boss, von Raumer, Muehlan.

Conducted experiments: Treiber, de Kanter, Roch, Gatfield, Boss, von Raumer, Muehlan, van Gerven.

Performed data analysis: Treiber, de Kanter, Roch, Gatfield, Boss, von Raumer, Muehlan.

Wrote or contributed to the writing of the manuscript: Treiber, de Kanter, Roch, Gatfield, Boss, Schindelholz, Jenck.

Footnotes

    • Received March 3, 2017.
    • Accepted June 20, 2017.
  • All experiments described in this report have been conducted in the research facilities of Actelion Pharmaceuticals Ltd. During manuscript preparation and review, Actelion Pharmaceuticals Ltd was acquired by Johnson & Johnson, and its drug discovery and early development activities subsequently transferred into a newly created company, Idorsia Pharmaceuticals Ltd. With the exception of Joop van Gerven, all authors of this report were employees of Idorsia Pharmaceuticals Ltd at the time of manuscript publication.

  • https://doi.org/10.1124/jpet.117.241596.

  • ↵Embedded ImageThis article has supplemental material available at jpet.aspetjournals.org.

Abbreviations

ACT-541468
(S)-(2-(5-chloro-4-methyl-1H-benzo[d]imidazol-2-yl)-2-methylpyrrolidin-1-yl)(5-methoxy-2-(2H-1,2,3-triazol-2-yl)phenyl)methanone
ACT-605143
(S)-(2-(5-chloro-1,4-dimethyl-1H-benzo[d]imidazol-2-yl)-2-methylpyrrolidin-1-yl)(5-methyl-2-(2H-1,2,3-triazol-2-yl)phenyl)methanone
ACT-658090
(S)-(2-(6-bromo-4-methyl-1H-benzo[d]imidazol-2-yl)-2-methylpyrrolidin-1-yl)(5-methyl-2-(pyrimidin-2-yl)phenyl)methanone
ADAM
advanced dissolution, absorption, and metabolism
AUC
area under the plasma concentration versus time curve
AUC0-last
area under the plasma concentration versus time curve up to the last measurable concentration
CL
total plasma clearance
CLint
intrinsic clearance
Cmax
peak observed plasma concentration
CRC
concentration-response curve
DMSO
dimethyl sulfoxide
DORA
dual orexin receptor antagonist
EEG
electroencephalogram, -graphic
EMG
electromyogram, -graphic
G418
(2R,3S,4R,5R,6S)-5-amino-6-[(1R,2S,3S,4R,6S)-4,6-diamino-3-[(2R,3R,4R,5R)-3,5-dihydroxy-5-methyl-4-methylaminooxan-2-yl]oxy-2-hydroxycyclohexyl]oxy- 2-(1-hydroxyethyl)oxane-3,4-diol
Kb
inhibition constant as determined by a functional assay
LC-MS/MS
liquid chromatography coupled to mass spectrometry
logD
distribution coefficient
MK-1064
5-(5-chloro-3-pyridyl)-N-((5,6-dimethoxy-2-pyridyl)methyl)-2-(2-pyridyl)pyridine-3-carboxamide
OX1
orexin-1 receptor
OX2
orexin-2 receptor
OxA
orexin A
PBPK-PD
physiology-based pharmacokinetic and pharmacodynamic (modeling)
PD
pharmacodynamic
PK
pharmacokinetic
REM
rapid eye movement
ROt1/2
receptor occupancy half-life
SB-649868
N-([(2S)-1-([5-(4-fluorophenyl)-2-methyl-4-thiazolyl]carbonyl)-2-piperidinyl]methyl)-4-benzofurancarboxamide
Tmax
time to reach peak plasma concentration
Vss
apparent volume of distribution at steady-state
σg
geometric standard deviation
  • Copyright © 2017 by The American Society for Pharmacology and Experimental Therapeutics

References

  1. ↵
    1. Aissaoui H,
    2. Koberstein R,
    3. Zumbrunn C,
    4. Gatfield J,
    5. Brisbare-Roch C,
    6. Jenck F,
    7. Treiber A, and
    8. Boss C
    (2008) N-Glycine-sulfonamides as potent dual orexin 1/orexin 2 receptor antagonists. Bioorg Med Chem Lett 18:5729–5733.
    OpenUrlPubMed
  2. ↵
    1. Baxter CA,
    2. Cleator E,
    3. Brands KMJ,
    4. Edwards JS,
    5. Reamer RA,
    6. Sheen FJ,
    7. Stewart GW,
    8. Strotman NA, and
    9. Wallace DJ
    (2011) The first large-scale synthesis of MK-4305: a dual orexin receptor antagonist for the treatment of sleep disorder. Org Process Res Dev 15:367–375.
    OpenUrl
  3. ↵
    1. Bettica P,
    2. Nucci G,
    3. Pyke C,
    4. Squassante L,
    5. Zamuner S,
    6. Ratti E,
    7. Gomeni R, and
    8. Alexander R
    (2012a) Phase I studies on the safety, tolerability, pharmacokinetics and pharmacodynamics of SB-649868, a novel dual orexin receptor antagonist. J Psychopharmacol 26:1058–1070.
    OpenUrlCrossRefPubMed
  4. ↵
    1. Bettica P,
    2. Squassante L,
    3. Groeger JA,
    4. Gennery B,
    5. Winsky-Sommerer R, and
    6. Dijk DJ
    (2012b) Differential effects of a dual orexin receptor antagonist (SB-649868) and zolpidem on sleep initiation and consolidation, SWS, REM sleep, and EEG power spectra in a model of situational insomnia. Neuropsychopharmacology 37:1224–1233.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Bonaventure P,
    2. Shelton J,
    3. Yun S,
    4. Nepomuceno D,
    5. Sutton S,
    6. Aluisio L,
    7. Fraser I,
    8. Lord B,
    9. Shoblock J,
    10. Welty N, et al.
    (2015a) Characterization of JNJ-42847922, a selective orexin-2 receptor antagonist, as a clinical candidate for the treatment of insomnia. J Pharmacol Exp Ther 354:471–482.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    1. Bonaventure P,
    2. Yun S,
    3. Johnson PL,
    4. Shekhar A,
    5. Fitz SD,
    6. Shireman BT,
    7. Lebold TP,
    8. Nepomuceno D,
    9. Lord B,
    10. Wennerholm M, et al.
    (2015b) A selective orexin-1 receptor antagonist attenuates stress-induced hyperarousal without hypnotic effects. J Pharmacol Exp Ther 352:590–601.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Boss C, Brotschi C, Gude M, Heidmann B, Sifferlen T, Von Raumer M, and Williams JT (2015a) inventors, Idorsia Pharmaceuticals Ltd, assignee. Crystalline form of (s)-(2-(6-chloro-7-methyl-1h-benzo[d]imidazol-2-yl)-2-methylpyrrolidin-1-yl)(5-methoxy-2-(2H-1,2,3-triazol-2-yl)phenyl)methanone and its use as orexin receptor antagonists. Publ. no. WO/2015/083070. International application number: PCT/IB2014/066508. 2015 Nov 6.
  8. ↵
    Boss C, Brotschi C, Gude M, Heidmann B, Sifferlen T, and Williams JT (2013) inventors, Idorsia Pharmaceuticals Ltd, assignee. Benzimidazole-proline derivatives. Publ. no. WO/2013/182972. International application number: PCT/IB2013/054567. 2013 Dec 12.
  9. ↵
    Boss C, Roch C, Brotschi C, Gude M, Heidmann B, Jenck F, Sifferlen T, Steiner MA, and Williams JT (2015b) inventors, Idorsia Pharmaceuticals Ltd, assignee. Use of benzimidazole-proline derivatives. Publ. no. WO/2015/083094. International appl. no. PCT/IB2014/066548. 2015 Nov 6.
  10. ↵
    1. Boss C,
    2. Roch C,
    3. Jenck F,
    4. Aissaoui H,
    5. Koberstein R,
    6. Sifferlen T, and
    7. Weller T
    (2008) Orexin receptor antagonism: a new principle in neuroscience. Chimia (Aarau) 62:974–979.
    OpenUrl
    1. Boss C,
    2. Roch-Brisbare C,
    3. Steiner MA,
    4. Treiber A,
    5. Dietrich H,
    6. Jenck F,
    7. von Raumer M,
    8. Sifferlen T,
    9. Brotschi C,
    10. Heidmann B, et al.
    (2014) Structure-activity relationship, biological, and pharmacological characterization of the proline sulfonamide ACT-462206: a potent, brain-penetrant dual orexin 1/orexin 2 receptor antagonist. ChemMedChem 9:2486–2496.
    OpenUrl
  11. ↵
    1. Carter ME,
    2. Schaich Borg J, and
    3. de Lecea L
    (2009) The brain hypocretins and their receptors: mediators of allostatic arousal. Curr Opin Pharmacol 9:39–45.
    OpenUrlCrossRefPubMed
  12. ↵
    1. Cheng Y and
    2. Prusoff WH
    (1973) Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol 22:3099–3108.
    OpenUrlCrossRefPubMed
  13. ↵
    1. Citrome L
    (2014) Suvorexant for insomnia: a systematic review of the efficacy and safety profile for this newly approved hypnotic - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int J Clin Pract 68:1429–1441.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Connor KM,
    2. Mahoney E,
    3. Jackson S,
    4. Hutzelmann J,
    5. Zhao X,
    6. Jia N,
    7. Snyder E,
    8. Snavely D,
    9. Michelson D,
    10. Roth T, et al.
    (2016) A phase II dose-ranging study evaluating the efficacy and safety of the orexin receptor antagonist filorexant (MK-6096) in patients with primary insomnia. Int J Neuropsychopharmacol 19:pyw022 DOI: 10.1093/ijnp/pyw022.
    OpenUrlCrossRefPubMed
  15. ↵
    1. Cox CD,
    2. Breslin MJ,
    3. Whitman DB,
    4. Schreier JD,
    5. McGaughey GB,
    6. Bogusky MJ,
    7. Roecker AJ,
    8. Mercer SP,
    9. Bednar RA,
    10. Lemaire W, et al.
    (2010) Discovery of the dual orexin receptor antagonist [(7R)-4-(5-chloro-1,3-benzoxazol-2-yl)-7-methyl-1,4-diazepan-1-yl][5-methyl-2-(2H-1,2,3-triazol-2-yl)phenyl]methanone (MK-4305) for the treatment of insomnia. J Med Chem 53:5320–5332.
    OpenUrlCrossRefPubMed
  16. ↵
    1. Cui D,
    2. Cabalu T,
    3. Yee KL,
    4. Small J,
    5. Li X,
    6. Liu B,
    7. Maciolek C,
    8. Smith S,
    9. Liu W,
    10. McCrea JB, et al.
    (2016) In vitro and in vivo characterisation of the metabolism and disposition of suvorexant in humans. Xenobiotica 46:882–895.
    OpenUrl
  17. ↵
    1. de Kanter R,
    2. Sidharta PN,
    3. Delahaye S,
    4. Gnerre C,
    5. Segrestaa J,
    6. Buchmann S,
    7. Kohl C, and
    8. Treiber A
    (2016) Physiologically-based pharmacokinetic modeling of macitentan: prediction of drug-drug interactions. Clin Pharmacokinet 55:369–380.
    OpenUrl
  18. ↵
    1. de Lecea L,
    2. Kilduff TS,
    3. Peyron C,
    4. Gao X,
    5. Foye PE,
    6. Danielson PE,
    7. Fukuhara C,
    8. Battenberg EL,
    9. Gautvik VT,
    10. Bartlett 2nd FS, et al.
    (1998) The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl Acad Sci USA 95:322–327.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Dingemanse J,
    2. Cruz HG,
    3. Gehin M, and
    4. Hoever P
    (2014) Pharmacokinetic interactions between the orexin receptor antagonist almorexant and the CYP3A4 inhibitors ketoconazole and diltiazem. J Pharm Sci 103:1548–1556.
    OpenUrl
  20. ↵
    1. Dugovic C,
    2. Shelton JE,
    3. Yun S,
    4. Bonaventure P,
    5. Shireman BT, and
    6. Lovenberg TW
    (2014) Orexin-1 receptor blockade dysregulates REM sleep in the presence of orexin-2 receptor antagonism. Front Neurosci 8:28 DOI: 10.3389/fnins.2014.00028.
    OpenUrlPubMed
  21. ↵
    1. Frey DJ,
    2. Ortega JD,
    3. Wiseman C,
    4. Farley CT, and
    5. Wright Jr KP
    (2011) Influence of zolpidem and sleep inertia on balance and cognition during nighttime awakening: a randomized placebo-controlled trial. J Am Geriatr Soc 59:73–81.
    OpenUrlCrossRefPubMed
  22. ↵
    1. Glass J,
    2. Lanctôt KL,
    3. Herrmann N,
    4. Sproule BA, and
    5. Busto UE
    (2005) Sedative hypnotics in older people with insomnia: meta-analysis of risks and benefits. BMJ 331:1169 10.1136/bmj.38623.768588.47.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Gotter AL,
    2. Forman MS,
    3. Harrell CM,
    4. Stevens J,
    5. Svetnik V,
    6. Yee KL,
    7. Li X,
    8. Roecker AJ,
    9. Fox SV,
    10. Tannenbaum PL, et al.
    (2016) Orexin 2 receptor antagonism is sufficient to promote NREM and REM sleep from mouse to man. Sci Rep 6:27147 10.1038/srep27147.
    OpenUrl
  24. ↵
    1. Gotter AL,
    2. Winrow CJ,
    3. Brunner J,
    4. Garson SL,
    5. Fox SV,
    6. Binns J,
    7. Harrell CM,
    8. Cui D,
    9. Yee KL,
    10. Stiteler M, et al.
    (2013) The duration of sleep promoting efficacy by dual orexin receptor antagonists is dependent upon receptor occupancy threshold. BMC Neurosci 14:90 10.1186/1471-2202-14-90.
    OpenUrlCrossRefPubMed
  25. ↵
    1. Gozzi A,
    2. Turrini G,
    3. Piccoli L,
    4. Massagrande M,
    5. Amantini D,
    6. Antolini M,
    7. Martinelli P,
    8. Cesari N,
    9. Montanari D,
    10. Tessari M, et al.
    (2011) Functional magnetic resonance imaging reveals different neural substrates for the effects of orexin-1 and orexin-2 receptor antagonists. PLoS One 6:e16406.
    OpenUrlCrossRefPubMed
  26. ↵
    1. Heidmann B,
    2. Gatfield J,
    3. Roch C,
    4. Treiber A,
    5. Tortoioli S,
    6. Brotschi C,
    7. Williams JT,
    8. Bolli MH,
    9. Abele S,
    10. Sifferlen T, et al.
    (2016) Discovery of highly potent dual orexin receptor antagonists via a scaffold-hopping approach. ChemMedChem 11:2132–2146 10.1002/cmdc.201600175.
    OpenUrl
  27. ↵
    1. Herring WJ,
    2. Snyder E,
    3. Budd K,
    4. Hutzelmann J,
    5. Snavely D,
    6. Liu K,
    7. Lines C,
    8. Roth T, and
    9. Michelson D
    (2012) Orexin receptor antagonism for treatment of insomnia: a randomized clinical trial of suvorexant. Neurology 79:2265–2274.
    OpenUrlCrossRefPubMed
  28. ↵
    1. Hoch M,
    2. Hoever P,
    3. Zisowsky J,
    4. Priestley A,
    5. Fleet D, and
    6. Dingemanse J
    (2012) Absolute oral bioavailability of almorexant, a dual orexin receptor antagonist, in healthy human subjects. Pharmacology 89:53–57.
    OpenUrlCrossRefPubMed
  29. ↵
    1. Hoever P,
    2. de Haas SL,
    3. Dorffner G,
    4. Chiossi E,
    5. van Gerven JM, and
    6. Dingemanse J
    (2012a) Orexin receptor antagonism: an ascending multiple-dose study with almorexant. J Psychopharmacol 26:1071–1080.
    OpenUrlCrossRefPubMed
  30. ↵
    1. Hoever P,
    2. Dorffner G,
    3. Beneš H,
    4. Penzel T,
    5. Danker-Hopfe H,
    6. Barbanoj MJ,
    7. Pillar G,
    8. Saletu B,
    9. Polo O,
    10. Kunz D, et al.
    (2012b) Orexin receptor antagonism, a new sleep-enabling paradigm: a proof-of-concept clinical trial. Clin Pharmacol Ther 91:975–985.
    OpenUrlCrossRefPubMed
  31. ↵
    1. Jacobson LH,
    2. Chen S,
    3. Mir S, and
    4. Hoyer D
    (2017) Orexin OX2 receptor antagonists as sleep aids. Curr Top Behav Neurosci 33:105–136.
    OpenUrl
  32. ↵
    1. Jamei M,
    2. Turner D,
    3. Yang J,
    4. Neuhoff S,
    5. Polak S,
    6. Rostami-Hodjegan A, and
    7. Tucker G
    (2009) Population-based mechanistic prediction of oral drug absorption. AAPS J 11:225–237.
    OpenUrlCrossRefPubMed
  33. ↵
    1. Johnson PL,
    2. Samuels BC,
    3. Fitz SD,
    4. Lightman SL,
    5. Lowry CA, and
    6. Shekhar A
    (2012) Activation of the orexin 1 receptor is a critical component of CO2-mediated anxiety and hypertension but not bradycardia. Neuropsychopharmacology 37:1911–1922.
    OpenUrlCrossRefPubMed
  34. ↵
    1. Jones HM,
    2. Gardner IB, and
    3. Watson KJ
    (2009) Modelling and PBPK simulation in drug discovery. AAPS J 11:155–166.
    OpenUrlCrossRefPubMed
  35. ↵
    1. Mahler SV,
    2. Moorman DE,
    3. Smith RJ,
    4. James MH, and
    5. Aston-Jones G
    (2014) Motivational activation: a unifying hypothesis of orexin/hypocretin function. Nat Neurosci 17:1298–1303.
    OpenUrlCrossRefPubMed
  36. ↵
    1. Mangion IK,
    2. Sherry BD,
    3. Yin J, and
    4. Fleitz FJ
    (2012) Enantioselective synthesis of a dual orexin receptor antagonist. Org Lett 14:3458–3461.
    OpenUrlPubMed
  37. ↵
    1. Merlo Pich E and
    2. Melotto S
    (2014) Orexin 1 receptor antagonists in compulsive behavior and anxiety: possible therapeutic use. Front Neurosci 8:26 DOI: 10.3389/fnins.2014.00026.
    OpenUrlPubMed
  38. ↵
    1. Mieda M,
    2. Hasegawa E,
    3. Kisanuki YY,
    4. Sinton CM,
    5. Yanagisawa M, and
    6. Sakurai T
    (2011) Differential roles of orexin receptor-1 and -2 in the regulation of non-REM and REM sleep. J Neurosci 31:6518–6526.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    1. Miller TR,
    2. Witte DG,
    3. Ireland LM,
    4. Kang CH,
    5. Roch JM,
    6. Masters JN,
    7. Esbenshade TA, and
    8. Hancock AA
    (1999) Analysis of apparent noncompetitive responses to competitive H(1)-histamine receptor antagonists in fluorescent imaging plate reader-based calcium assays. J Biomol Screen 4:249–258.
    OpenUrlCrossRefPubMed
  40. ↵
    1. Muehlan C,
    2. Juif PE,
    3. Van Gerven J,
    4. Heuberger J, and
    5. Dingemanse J
    (2017) First-in-man study of ACT-541468, a novel dual orexin receptor antagonist: characterization of its pharmacokinetics, pharmacodynamics, safety and tolerability. In: American Society for Clinical Pharmacology and Therapeutics 2017 Annual Meeting Abstracts. Clin Pharmacol Ther 101:S5–S99.
    OpenUrl
  41. ↵
    1. Otmani S,
    2. Demazières A,
    3. Staner C,
    4. Jacob N,
    5. Nir T,
    6. Zisapel N, and
    7. Staner L
    (2008) Effects of prolonged-release melatonin, zolpidem, and their combination on psychomotor functions, memory recall, and driving skills in healthy middle aged and elderly volunteers. Hum Psychopharmacol 23:693–705.
    OpenUrlCrossRefPubMed
  42. ↵
    1. Peyron C,
    2. Faraco J,
    3. Rogers W,
    4. Ripley B,
    5. Overeem S,
    6. Charnay Y,
    7. Nevsimalova S,
    8. Aldrich M,
    9. Reynolds D,
    10. Albin R, et al.
    (2000) A mutation in a case of early onset narcolepsy and a generalized absence of hypocretin peptides in human narcoleptic brains. Nat Med 6:991–997.
    OpenUrlCrossRefPubMed
  43. ↵
    1. Ramirez AD,
    2. Gotter AL,
    3. Fox SV,
    4. Tannenbaum PL,
    5. Yao L,
    6. Tye SJ,
    7. McDonald T,
    8. Brunner J,
    9. Garson SL,
    10. Reiss DR, et al.
    (2013) Dual orexin receptor antagonists show distinct effects on locomotor performance, ethanol interaction and sleep architecture relative to gamma-aminobutyric acid-A receptor modulators. Front Neurosci 7:254 DOI: 10.3389/fnins.2013.00254.
    OpenUrlPubMed
  44. ↵
    1. Rodgers T and
    2. Rowland M
    (2007) Mechanistic approaches to volume of distribution predictions: understanding the processes. Pharm Res 24:918–933.
    OpenUrlCrossRefPubMed
  45. ↵
    1. Sakurai T
    (1999) Orexins and orexin receptors: implication in feeding behavior. Regul Pept 85:25–30.
    OpenUrlCrossRefPubMed
  46. ↵
    1. Sakurai T
    (2007) The neural circuit of orexin (hypocretin): maintaining sleep and wakefulness. Nat Rev Neurosci 8:171–181.
    OpenUrlCrossRefPubMed
    1. Sifferlen T,
    2. Boller A,
    3. Chardonneau A,
    4. Cottreel E,
    5. Gatfield J,
    6. Treiber A,
    7. Roch C,
    8. Jenck F,
    9. Aissaoui H,
    10. Williams JT, et al.
    (2015) Substituted pyrrolidin-2-ones: Centrally acting orexin receptor antagonists promoting sleep. Part 2. Bioorg Med Chem Lett 25:1884–1891.
    OpenUrl
  47. ↵
    1. Sifferlen T,
    2. Boss C,
    3. Cottreel E,
    4. Koberstein R,
    5. Gude M,
    6. Aissaoui H,
    7. Weller T,
    8. Gatfield J,
    9. Brisbare-Roch C, and
    10. Jenck F
    (2010) Novel pyrazolo-tetrahydropyridines as potent orexin receptor antagonists. Bioorg Med Chem Lett 20:1539–1542.
    OpenUrlPubMed
    1. Sifferlen T,
    2. Koberstein R,
    3. Cottreel E,
    4. Boller A,
    5. Weller T,
    6. Gatfield J,
    7. Brisbare-Roch C,
    8. Jenck F, and
    9. Boss C
    (2013) Synthesis, structure-activity relationship studies, and identification of novel 5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine derivatives as dual orexin receptor antagonists. Part 1. Bioorg Med Chem Lett 23:2212–2216.
    OpenUrl
  48. ↵
    1. Sun H,
    2. Kennedy WP,
    3. Wilbraham D,
    4. Lewis N,
    5. Calder N,
    6. Li X,
    7. Ma J,
    8. Yee KL,
    9. Ermlich S,
    10. Mangin E, et al.
    (2013) Effects of suvorexant, an orexin receptor antagonist, on sleep parameters as measured by polysomnography in healthy men. Sleep 36:259–267.
    OpenUrlPubMed
  49. ↵
    1. Sun H,
    2. Yee KL,
    3. Gill S,
    4. Liu W,
    5. Li X,
    6. Panebianco D,
    7. Mangin E,
    8. Morrison D,
    9. McCrea J,
    10. Wagner JA, et al.
    (2015) Psychomotor effects, pharmacokinetics and safety of the orexin receptor antagonist suvorexant administered in combination with alcohol in healthy subjects. J Psychopharmacol 29:1159–1169.
    OpenUrlCrossRefPubMed
  50. ↵
    1. Tannenbaum PL,
    2. Tye SJ,
    3. Stevens J,
    4. Gotter AL,
    5. Fox SV,
    6. Savitz AT,
    7. Coleman PJ,
    8. Uslaner JM,
    9. Kuduk SD,
    10. Hargreaves R, et al.
    (2016) Inhibition of orexin signaling promotes sleep yet preserves salient arousability in monkeys. Sleep 39:603–612.
    OpenUrl
  51. ↵
    TGA (2014) Extract from the clinical evaluation report for suvorexant. Available at: www.tga.gov.au/sites/default/files/auspar-suvorexant-150411-cer.pdf. Accessed on: November 11, 2016. Australian Government, Department of Health, Therapeutic Goods Administration.
  52. ↵
    1. Thannickal TC,
    2. Moore RY,
    3. Nienhuis R,
    4. Ramanathan L,
    5. Gulyani S,
    6. Aldrich M,
    7. Cornford M, and
    8. Siegel JM
    (2000) Reduced number of hypocretin neurons in human narcolepsy. Neuron 27:469–474.
    OpenUrlCrossRefPubMed
  53. ↵
    1. Tsujino N and
    2. Sakurai T
    (2009) Orexin/hypocretin: a neuropeptide at the interface of sleep, energy homeostasis, and reward system. Pharmacol Rev 61:162–176.
    OpenUrlAbstract/FREE Full Text
  54. ↵
    1. Vermeeren A,
    2. Sun H,
    3. Vuurman EF,
    4. Jongen S,
    5. Van Leeuwen CJ,
    6. Van Oers AC,
    7. Palcza J,
    8. Li X,
    9. Laethem T,
    10. Heirman I, et al.
    (2015) On-the-road driving performance the morning after bedtime use of suvorexant 20 and 40 mg: a study in non-elderly healthy volunteers. Sleep 38:1803–1813.
    OpenUrl
  55. ↵
    1. Verzijl GKM,
    2. de Vries AHM,
    3. de Vries JG,
    4. Kapitan P,
    5. Dax T,
    6. Helms M,
    7. Nazir Z,
    8. Skranc W,
    9. Imboden C,
    10. Stichler J, et al.
    (2013) Catalytic asymmetric reduction of a 3,4-dihydroisoquinoline for the large-scale production of almorexant: hydrogenation or transfer hydrogenation? Org Process Res Dev 17:1531–1539.
    OpenUrl
  56. ↵
    1. Winrow CJ,
    2. Gotter AL,
    3. Cox CD,
    4. Tannenbaum PL,
    5. Garson SL,
    6. Doran SM,
    7. Breslin MJ,
    8. Schreier JD,
    9. Fox SV,
    10. Harrell CM, et al.
    (2012) Pharmacological characterization of MK-6096 - a dual orexin receptor antagonist for insomnia. Neuropharmacology 62:978–987.
    OpenUrlCrossRefPubMed
  57. ↵
    1. Yoshida Y,
    2. Naoe Y,
    3. Terauchi T,
    4. Ozaki F,
    5. Doko T,
    6. Takemura A,
    7. Tanaka T,
    8. Sorimachi K,
    9. Beuckmann CT,
    10. Suzuki M, et al.
    (2015) Discovery of (1R,2S)-2-[(2,4-Dimethylpyrimidin-5-yl)oxy]methyl-2-(3-fluorophenyl)-N-(5-fluoropyridin-2-yl)cyclopropanecarboxamide (E2006): A Potent and Efficacious Oral Orexin Receptor Antagonist. J Med Chem 58:4648–4664.
    OpenUrl
  58. ↵
    1. Zammit GK
    (2007) The prevalence, morbidities, and treatments of insomnia. CNS Neurol Disord Drug Targets 6:3–16.
    OpenUrlPubMed
PreviousNext
Back to top

In this issue

Journal of Pharmacology and Experimental Therapeutics: 362 (3)
Journal of Pharmacology and Experimental Therapeutics
Vol. 362, Issue 3
1 Sep 2017
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Journal of Pharmacology and Experimental Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
The Use of Physiology-Based Pharmacokinetic and Pharmacodynamic Modeling in the Discovery of the Dual Orexin Receptor Antagonist ACT-541468
(Your Name) has forwarded a page to you from Journal of Pharmacology and Experimental Therapeutics
(Your Name) thought you would be interested in this article in Journal of Pharmacology and Experimental Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Research ArticleDrug Discovery and Translational Medicine

Role of Physiology-Based Modeling in ACT-541468 Discovery

Alexander Treiber, Ruben de Kanter, Catherine Roch, John Gatfield, Christoph Boss, Markus von Raumer, Benno Schindelholz, Clemens Muehlan, Joop van Gerven and Francois Jenck
Journal of Pharmacology and Experimental Therapeutics September 1, 2017, 362 (3) 489-503; DOI: https://doi.org/10.1124/jpet.117.241596

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Research ArticleDrug Discovery and Translational Medicine

Role of Physiology-Based Modeling in ACT-541468 Discovery

Alexander Treiber, Ruben de Kanter, Catherine Roch, John Gatfield, Christoph Boss, Markus von Raumer, Benno Schindelholz, Clemens Muehlan, Joop van Gerven and Francois Jenck
Journal of Pharmacology and Experimental Therapeutics September 1, 2017, 362 (3) 489-503; DOI: https://doi.org/10.1124/jpet.117.241596
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Authorship Contributions
    • Footnotes
    • Abbreviations
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF + SI
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Metformin alters lipidome independent of diabetes control
  • PK/PD relationship of RyR2 inhibitor ent-verticilide
  • SOS1 and MEK inhibitors combination therapy in neurofibroma
Show more Drug Discovery and Translational Medicine

Similar Articles

Advertisement
  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About JPET
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Drug Metabolism and Disposition
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-0103 (Online)

Copyright © 2023 by the American Society for Pharmacology and Experimental Therapeutics