throbber
Downloaded from
`
`molpharm.aspetjournals.org
`
` at ASPET Journals on January 13, 2016
`
`1521-0111/12/8202-226–235$25.00
`MOLECULAR PHARMACOLOGY
`Copyright © 2012 The American Society for Pharmacology and Experimental Therapeutics
`Mol Pharmacol 82:226–235, 2012
`
`Vol. 82, No. 2
`78154/3781922
`
`␥-Hydroxybutyrate (GHB)-Induced Respiratory Depression:
`Combined Receptor-Transporter Inhibition Therapy for
`Treatment in GHB Overdose
`
`Bridget L. Morse, Nisha Vijay, and Marilyn E. Morris
`Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State
`University of New York, Buffalo, New York
`Received February 7, 2012; accepted May 4, 2012
`
`ABSTRACT
`Overdose of ␥-hydroxybutyrate (GHB) frequently causes respi-
`ratory depression, occasionally resulting in death; however,
`little is known about the dose-response relationship or effects
`of potential overdose treatment strategies on GHB-induced
`respiratory depression.
`In these studies, the parameters of
`respiratory rate, tidal volume, and minute volume were mea-
`sured using whole-body plethysmography in rats administered
`GHB. Intravenous doses of 200, 600, and 1500 mg/kg were
`administered to assess the dose-dependent effects of GHB on
`respiration.To determine the receptors involved in GHB-induced
`respiratory depression, a specific GABAB receptor antagonist,
`(2S)-(⫹)-5,5-dimethyl-2-morpholineacetic acid (SCH50911), and a
`specific GABAA receptor antagonist, bicuculline, were adminis-
`tered before GHB. The potential therapeutic strategies of receptor
`inhibition and monocarboxylate transporter (MCT) inhibition were
`assessed by inhibitor administration 5 min after GHB. The primary
`
`effect of GHB on respiration was a dose-dependent decrease in
`respiratory rate, accompanied by an increase in tidal volume,
`resulting in little change in minute volume. Pretreatment with 150
`mg/kg SCH50911 completely prevented the decrease in respira-
`tory rate, indicating agonism at GABAB receptors to be primarily
`responsible for GHB-induced respiratory depression. Administra-
`tion of 50 mg/kg SCH50911 after GHB completely reversed the
`decrease in respiratory rate; lower doses had partial effects. Ad-
`ministration of the MCT inhibitor L-lactate increased GHB renal
`and total clearance, also improving respiratory rate. Administra-
`tion of 5 mg/kg SCH50911 plus L-lactate further improved respi-
`ratory rate compared with the same dose of either agent alone,
`indicating that GABAB and MCT inhibitors, alone and in combina-
`tion, represent potential treatment options for GHB-induced re-
`spiratory depression.
`
`Introduction
`␥-Hydroxybutyate (GHB) is a short-chain fatty acid pres-
`ent endogenously in many human tissues, resulting from
`production via GABA metabolism (Maitre, 1997). GHB has
`also recently been identified as a useful therapeutic agent for
`the treatment of narcolepsy and excessive daytime sleepiness
`in the form of sodium oxybate (Xyrem; Jazz Pharmaceuticals,
`Palo Alto, CA). However, GHB has become more popularly
`
`This work was supported by the National Institutes of Health National
`Institute on Drug Abuse [Grant DA023223] and Pfizer Global Research and
`Development.
`This work was previously presented in part as an abstract: Morse B,
`Uhlander J, and Morris M. Respiratory depression in ␥-hydroxybutyrate over-
`dose: interaction with ethanol and treatment using monocarboxylate trans-
`porter inhibition, AAPS Annual Meeting and Exposition; 2011 Oct 23–27; Wash-
`ington DC. American Association of Pharmaceutical Sciences, Arlington, VA.
`Article, publication date, and citation information can be found at
`http://molpharm.aspetjournals.org.
`http://dx.doi.org/10.1124/mol.112.078154.
`
`known as a drug of abuse. According to reports from the Drug
`Abuse Warning Network, there have consistently been 1000
`to 2000 GHB-related emergency department visits reported
`annually in the United States over the past several years
`(Substance Abuse and Mental Health Services Administra-
`tion, 2011). GHB overdose can result in manifestations in-
`cluding sedation, coma, hypothermia, bradycardia, respira-
`tory depression, and death (Li et al., 1998; Sporer et al., 2003;
`Caldicott et al., 2004; Galicia et al., 2011). In a recent report
`of known GHB-associated fatalities, the most common cause
`of mortality was cardiorespiratory arrest (Zvosec et al.,
`2011). Respiratory depression with the need for mechanical
`ventilation is also frequently reported in nonfatal cases of
`GHB intoxication (Li et al., 1998; Mason and Kerns, 2002;
`Liechti and Kupferschmidt, 2004).
`Although respiratory depression is a common symptom of
`GHB overdose, neither the dose-dependent effects of GHB on
`this measure nor the neurotransmitter receptors involved in
`
`ABBREVIATIONS: GHB, ␥-hydroxybutyrate; MCT, monocarboxylate transporter; SCH50911, (2S)-(⫹)-5,5-dimethyl-2-morpholineacetic acid; LC,
`liquid chromatography; MS/MS, mass spectrometry; Emax, maximum effect; Td, duration of effect; AUC, area under the plasma concentration-time
`curve; ABEC, area below the effect curve.
`
`226
`
`Page 1 of 10
`
`JAZZ EXHIBIT 2004
`Ranbaxy Inc. (Petitioner) v. Jazz Pharms. Ireland Ltd. (Patent Owner)
`Case IPR2016-00024
`
`

`
`Downloaded from
`
`molpharm.aspetjournals.org
`
` at ASPET Journals on January 13, 2016
`
`GHB-induced respiratory depression have been investigated.
`There are several proposed actions of GHB, including 1)
`direct action at GABAB receptors (Bernasconi et al., 1992), 2)
`direct action at its own putative GHB receptor (Maitre,
`1997), and 3) indirect action at GABA receptors via GABA
`production/release (Hechler et al., 1997; Gobaille et al.,
`1999). Although evidence exists for each of these mechanisms
`in vitro and/or in vivo, many of the toxicological effects of
`GHB, including sedation, hypothermia, and fatality, can be
`attributed to agonism at GABAB receptors (Carai et al., 2001,
`2005; Kaupmann et al., 2003).
`Along with a complex pharmacologic profile, the pharma-
`cokinetics of GHB are also notably complicated. In humans,
`GHB exhibits dose-dependent pharmacokinetics, even at
`therapeutic concentrations (Palatini et al., 1993). Rats simi-
`larly display nonlinear pharmacokinetics, due to several con-
`centration-dependent processes including saturable oral absorp-
`tion, saturable metabolism, and saturable renal reabsorption
`(Lettieri and Fung, 1979; Morris et al., 2005). In both humans and
`rats, GHB metabolism is the predominant route of elimination at
`low doses, and renal excretion of unchanged drug is minimal
`(Lettieri and Fung, 1976; Brenneisen et al., 2004). Although lim-
`ited information exists for supratherapeutic GHB doses in hu-
`mans, it has been well documented in rats that renal clearance
`becomes an increasingly important route of elimination as GHB
`doses are increased (Morris et al., 2005). This nonlinear renal
`clearance can be attributed to a concentration-dependent trans-
`port process, leading to saturable renal reabsorption, demon-
`strated in our laboratory to involve the group of transporters
`known as monocarboxylate transporters (MCTs) (Morris et al.,
`2005; Wang et al., 2006). MCTs are proton-dependent transporters
`expressed throughout the body, and GHB is an identified sub-
`strate for MCTs 1, 2, and 4 (Wang et al., 2006; Wang and Morris,
`2007). The ubiquitous expression of these transporters includes
`that in the intestine, kidney, and brain, regions of interest regard-
`ing GHB pharmacokinetics. Because of their role in the renal
`reabsorption of GHB, inhibition of these transporters represents a
`potential therapeutic strategy for GHB overdose. This strategy has
`been demonstrated to translate to in vivo effects on GHB disposi-
`tion, and administration of MCT inhibitors increases the renal and
`total clearance in animal models of GHB overdose (Morris et al.,
`2005; Wang et al., 2008a,b). Likewise, the administration of the
`MCT inhibitor L-lactate, in combination with osmotic diuresis,
`increases the renal clearance of GHB in humans, as demonstrated
`in our pilot clinical study (Morris et al., 2011).
`The first aim of this research was to investigate the dose-
`response relationship of GHB-induced respiratory depres-
`sion, including the primary neurotransmitter receptors in-
`volved in eliciting this effect. The second was to assess
`potential treatment strategies, including MCT and receptor
`inhibition, for improving GHB-induced respiratory depres-
`sion, because the application of these strategies for treating
`this pharmacodynamic endpoint have not been evaluated
`previously.
`
`Materials and Methods
`Chemicals and Reagents. Sodium GHB used in these studies
`was provided by the National Institute on Drug Abuse. Deuterated
`GHB (GHB-d6) was purchased from Cerilliant Corporation (Round
`Rock, TX). Sodium L-lactate and bicuculline methiodide were pur-
`chased from Sigma-Aldrich (St. Louis, MO). (2S)-(⫹)-5,5-Dimethyl-
`
`GHB-Induced Respiratory Depression
`
`227
`
`2-morpholineacetic acid (SCH50911) was purchased from Tocris Bio-
`science (Ellisville, MO). High-performance liquid chromatography-
`grade acetonitrile and acetic acid were purchased from Honeywell
`Burdick & Jackson (Muskegon, MI).
`Animals and Animal Surgery. Male Sprague-Dawley rats (Har-
`lan, Indianapolis, IN) weighing 270 to 330 g were used for all exper-
`iments. Animals were housed under controlled temperature and
`humidity with an artificial 12-h light/dark cycle, and food was avail-
`able ad libitum. All animal protocols were approved by the Institu-
`tional Animal Care and Use Committee at the University at Buffalo.
`Animals were allowed to acclimate to their environment for a mini-
`mum of 1 week before surgical implantation of jugular and femoral
`vein cannulae under anesthesia with ketamine-xylazine. Cannulae
`were flushed daily with 40 IU/ml heparinized saline to maintain
`patency. Animals were allowed a minimum of 72 h for recovery from
`surgery before drug administration.
`Plethysmography. Measurement of respiration in these studies
`was performed using a whole-body plethysmograph (model PLY4213;
`Buxco Research Systems, Wilmington, NC). Plethysmography equip-
`ment included unrestrained plethysmography chambers consisting
`of a main (animal) chamber and reference chamber for buffering
`changes in atmospheric pressure. The plethysmography chambers
`were connected to a Rodent Bias Flow Supply (BFL0250) to draw
`expired CO2 out of the chambers and provide a smoothed flow of
`room air at a flow rate of 2.5 l/min per chamber. The plethysmogra-
`phy chambers included ports to which a pressure sensor was con-
`nected and led to the MAX 1500 preamplifier. Signals were collected,
`visualized, and quantitated using BioSystem XA software. Two ad-
`ditional ports were included in the chambers for the insertion of
`jugular and femoral vein cannulae, allowing for drug administration
`and blood sampling. Urine was collected at the base of the chamber
`at intervals by opening an additional port at the base. Calibration of
`chamber pressure was performed before every experiment by injec-
`tion of 5 ml of air through the base port. At each recording, signals
`were collected for six intervals of 10 s each and averaged to represent
`1 min of recording. Measurements for the parameters of respiratory
`frequency (rate), tidal volume, and minute volume (rate 䡠 tidal vol-
`ume) were quantitated for each recording.
`Pharmacokinetic/Pharmacodynamic Studies. Rats were
`placed in plethysmography chambers 1 h before drug administration
`and allowed to acclimate to the chambers for 45 min before five
`baseline measurements of 1 min each were collected over 15 min. In
`all studies, GHB administration was considered time 0, and respi-
`ration measurements were recorded at 2.5, 5, 7.5, 10, 15, 20, 25, and
`30 min and every 15 min thereafter for 480 min. Blood samples were
`collected, and collection times were optimized for each GHB dose
`according to previous studies (Felmlee et al., 2010b, 2011). Urine was
`collected at intervals up to 480 min. For overlapping pharmacoki-
`netic/pharmacodynamic time points, blood and urine samples were
`taken directly after the recording of respiratory measurements.
`Dose-Dependent Effects of GHB on Respiration. To assess
`the dose-response relationship of GHB-induced respiratory depres-
`sion, rats were administered GHB intravenously in doses of 200, 600,
`and 1500 mg/kg (four to six animals per dose). GHB was injected over
`1 to 2 min as a 300 mg/ml solution in sterile water via the jugular
`vein cannula. A placebo control group received a 5 ml/kg saline bolus.
`Neurotransmitter Receptors Involved in GHB-Induced Re-
`spiratory Depression. To determine the primary receptors in-
`volved in GHB-induced respiratory depression, rats were pretreated
`with specific receptor antagonists. Bicuculline methiodide (5 mg/kg)
`was administered for inhibition of GABAA receptors and SCH50911
`(150 mg/kg) for inhibition of GABAB receptors (three to four animals
`per group). Inhibitors were administered immediately after the col-
`lection of baseline respiratory measurements and 1500 mg/kg GHB
`was administered 5 min later. Data from dose-dependent experi-
`ments were used as the control. Bicuculline methiodide was admin-
`istered as a 5 mg/ml solution in saline and SCH50911 as a 50 mg/ml
`solution in saline via the jugular vein cannula.
`
`Page 2 of 10
`
`

`
`Downloaded from
`
`molpharm.aspetjournals.org
`
` at ASPET Journals on January 13, 2016
`
`228
`
`Morse et al.
`
`Potential Treatment Strategies. To assess the effect of poten-
`tial treatment strategies on GHB-induced respiratory depression,
`treatments were administered intravenously 5 min after 1500 mg/kg
`GHB. Treatment strategies included SCH50911 (2.5, 5, 10, and 50
`mg/kg), the MCT inhibitor L-lactate (66 mg/kg bolus followed by a
`302.5 mg/kg/h infusion for 8 h), and combination therapy of 5 mg/kg
`SCH50911 plus the same dose of L-lactate. Treatment groups in-
`cluded three to five animals per group, and were compared with the
`1500 mg/kg control group from dose-dependent experiments to de-
`termine the effects of treatment on GHB-induced respiratory depres-
`sion. The same L-lactate dose was also administered alone at time 0
`to assess potential effects of this agent on respiration. In these
`experiments, SCH50911 was administered as a 2.5, 5, 10, or 50
`mg/ml solution in saline via the jugular vein cannula and L-lactate as
`a 40 mg/ml solution in sterile water via the femoral vein cannula.
`Plasma and Urine Sample Analysis. GHB plasma concentra-
`tions were determined using an LC-MS/MS method, similar to those
`published previously (Fung et al., 2008; Felmlee et al., 2010a).
`Plasma samples were prepared by adding 5 ␮l of GHB-d6 (125 ␮g/ml)
`to 50 ␮l of sample. Plasma standards and quality controls were
`prepared by adding 5 ␮l of GHB-d6 and 5 ␮l of GHB stock solution to
`45 ␮l of blank plasma, and 800 ␮l of 0.1% formic acid in acetonitrile
`was added to precipitate the plasma proteins. The samples were
`vortexed, followed by centrifugation at 10,000g for 20 min at 4°C.
`Then 750 ␮l of the supernatant was aspirated and evaporated under
`a stream of nitrogen gas. The samples were reconstituted in 250 ␮l of
`aqueous mobile phase.
`The LC-MS/MS assay was performed on an Agilent 1100 series
`high-performance liquid chromatography system with binary pump
`and autosampler (Agilent Technologies, Santa Clara, CA) connected
`to a PerkinElmer Sciex API 3000 triple quadrupole tandem mass
`spectrometer with a TurboIonSpray (Applied Biosystems, Foster
`City, CA). Chromatographic separation was achieved by injecting 7
`␮l of sample on an Xterra MS C18 column (250 ⫻ 2.1 mm i.d., 5-␮m
`particle size; Waters, Milford, MA). Mobile phase A consisted of 5:95
`acetonitrile-water with 0.1% acetic acid and mobile phase B con-
`sisted of 95:5 acetonitrile-water with 0.1% acetic acid. The flow rate
`was 200 ␮l/min with the following gradient elution profile: 100 to
`68% A over 7 min; 68 to 10% A over 3 min; and 10 to 100% over 5 min
`for a total run time of 15 min. The mass spectrometer was operated
`in a positive ionization mode with multiple reaction monitoring.
`Q1/Q3 m/z ratios for the parent/product ions of GHB and GHB-d6
`were 105.2/87.2 and 111.1/93.2, respectively. The mass spectrometer
`parameters were optimized at a declustering potential of 18 V, fo-
`cusing potential of 100 V, collision energy of 20 V, entrance potential
`of 10 V, and collision cell exit potential of 5 V. The ion spray voltage
`was set at 5500 V with temperature at 350°C. Nebulizer and curtain
`gas flow were set at 10 and 8 ml/min, respectively. The retention
`time for GHB was 4.15 min. The data were analyzed using Analyst
`software version 1.4.2 (Applied Biosystems).
`Regression analysis of peak area ratios of GHB/GHB-d6 to GHB
`concentrations was used to assess linearity of the curve. The intra-
`day and interday precision and accuracy were determined using
`quality control (QC) samples at 10 ␮g/ml (low QC), 125 ␮g/ml (me-
`dium QC), and 400 ␮g/ml (high QC). For determination of the intra-
`day precision and accuracy, quality control samples were analyzed in
`triplicate on each day, whereas for the interday precision and accu-
`racy, quality control samples were analyzed on three different days.
`A calibration curve was run on each analysis day along with the
`quality controls. The precision was determined by the coefficient of
`variation, and accuracy was measured by comparing the calculated
`concentration with the known concentration.
`Urine samples were prepared and analyzed for GHB using a
`previously described LC-MS/MS method (Felmlee et al., 2010b).
`Plasma lactate concentrations were determined using a YSI 1500
`Sport Lactate Analyzer (YSI, Inc., Yellow Springs, OH).
`Data and Statistical Analysis. Pharmacokinetic parameters
`were determined via noncompartmental analysis using WinNonlin
`
`5.2 (Pharsight, Mountain View, CA). The area below the plasma
`concentration-time curve (AUC) was determined using the trapezoi-
`dal method. Total clearance (Cl) was determined as dose/AUC. Renal
`clearance (ClR) was determined as Ae/AUC, where Ae represents the
`amount excreted in the urine. Percentage of urinary excretion was
`calculated as Ae/dose. Metabolic or nonrenal clearance (Clm) was
`calculated as Cl ⫺ ClR. The pharmacodynamic descriptors of area
`below the effect curve (ABEC), maximum effect (Emax), time of max-
`imum effect (Tmax), and duration of effect (Td) were used to deter-
`mine the effects of inhibitor administration on GHB-induced respi-
`ratory depression. ABEC was calculated using WinNonlin. Td was
`determined for each animal as the time to return to its individual
`baseline respiratory frequency. Statistical analysis was performed
`using SigmaPlot 10.0 (Systat Software, Inc., San Jose, CA). Differ-
`ences were considered significant when p ⬍ 0.05. One-way analysis
`of variance followed by Dunnett’s or Tukey’s post hoc tests was used
`to determine statistically significant differences in mean pharmaco-
`kinetic and pharmacodynamic parameters between groups. Paired t
`tests were used to determine statistically significant changes in
`respiratory parameters compared with baseline. In determining the
`effects of L-lactate alone on respiration, the average of the last hour
`of respiratory measurements was compared with the individual av-
`erage baseline values. Mean steady-state lactate plasma concentra-
`tions were calculated as the average of hourly values beginning at 60
`min.
`
`Results
`Plasma GHB LC-MS/MS Assay. The lower limit of
`quantification for GHB in plasma was found to be 5 ␮g/ml
`with acceptable error in precision and accuracy of less than
`20%. The endogenous concentrations of GHB in plasma are
`negligible compared with GHB concentrations obtained
`after administration of the lowest dose in our studies
`(Fung et al., 2004); therefore, the endogenous concentra-
`tions were not included in the calculation of GHB concen-
`trations in plasma. The standard curve for GHB ranged
`from 5 to 500 ␮g/ml based on regression analysis of peak
`area ratios of GHB/GHB-d6 to GHB concentrations with a
`correlation coefficient (r2 ⬎0.999). The intraday and inter-
`
`TABLE 1
`Intraday and interday accuracy and precision for GHB in rat plasma
`Each measured concentration is the mean of triplicate measurements. The analysis
`was performed over 3 days.
`
`Nominal
`Concentration
`
`Measured
`Concentration
`
`S.D.
`
`Precision
`
`Accuracy
`
`Intraday
`
`Interday
`
`␮g/ml
`
`10.8
`121
`375
`10.5
`118
`368
`
`0.12
`4.36
`6.93
`0.32
`2.96
`6.38
`
`CV%
`1.07
`3.60
`1.85
`3.05
`2.50
`1.73
`
`%
`107.7
`96.8
`93.7
`105.2
`94.9
`92.0
`
`10
`125
`400
`10
`125
`400
`
`CV, coefficient of variation.
`
`TABLE 2
`Nonlinear pharmacokinetics of GHB
`GHB was administered intravenously. Data are presented as mean (S.D.); n ⫽ 4 to
`6. One-way analysis of variance followed by Tukey’s post hoc test was used to
`determine statistically significant differences in pharmacokinetic parameters.
`
`200 mg/kg
`
`600 mg/kg
`
`1500 mg/kg
`
`Cl, ml 䡠 kg⫺1 䡠 min⫺1
`6.00 (0.74)a
`7.60 (0.29)
`ClR, ml䡠 kg⫺1 䡠 min⫺1
`1.68 (0.75)a
`0.444 (0.20)
`26.7 (11)a
`6.0 (3)
`Urinary excretion, %
`a Significantly different from 200 mg/kg GHB (P ⬍ 0.05).
`b Significantly different from 200 and 600 mg/kg GHB (P ⬍ 0.05).
`
`5.16 (0.70)a
`3.18 (0.66)a,b
`60.1 (7)a,b
`
`Page 3 of 10
`
`

`
`GHB-Induced Respiratory Depression
`
`229
`
`Downloaded from
`
`molpharm.aspetjournals.org
`
` at ASPET Journals on January 13, 2016
`
`Fig. 1. Dose-dependent effects of GHB
`on measures of respiration. GHB was
`administered intravenously at time 0.
`Data are presented as mean ⫾ S.D.;
`n ⫽ 4 to 6.
`
`Page 4 of 10
`
`

`
`Downloaded from
`
`molpharm.aspetjournals.org
`
` at ASPET Journals on January 13, 2016
`
`Fig. 2. Effect of GHB administration
`on respiratory pattern. Displayed are
`sample 10-s interval plethysmogra-
`phy traces obtained at baseline (A)
`and 30 min after administration of
`GHB 1500 mg/kg i.v. (B).
`
`TABLE 3
`Effects of specific receptor antagonists on the
`pharmacokinetics/pharmacodynamics of GHB (1500 mg/kg i.v.)
`SCH50911 (150 mg/kg) and bicuculline methiodide (5 mg/kg) were administered
`intravenously 5 min before GHB. Data are presented as mean (S.D.); n ⫽ 3 to 5.
`One-way analysis of variance followed by Dunnett’s post hoc test was used to
`determine statistically significant differences in mean pharmacokinetic and phar-
`macodynamic parameters with inhibitor administration compared with those with
`GHB alone.
`
`GHB
`
`GHB ⫹
`SCH50911
`
`GHB ⫹ Bicuc-
`ulline
`
`5.16 (0.70)
`3.18 (0.66)
`1.99 (0.17)
`10,500 (2700)
`
`6.07 (0.47)
`2.84 (0.32)
`3.23 (0.78)*
`—a
`
`5.02 (0.14)
`2.72 (0.78)
`2.30 (0.63)
`10,900 (1300)
`
`15 (9)
`
`67.5 (13)
`
`Cl, ml 䡠 kg⫺1 䡠 min⫺1
`ClR, ml䡠 kg⫺1 䡠 min⫺1
`Clm, ml䡠 kg⫺1 䡠 min⫺1
`Frequency ABEC,
`breaths
`Frequency Emax,
`breaths/min
`Frequency Tmax, min
`* Significantly different from GHB alone (P ⬍ 0.05).
`a —, no ABEC, Emax, orT max values could be calculated because respiration is
`similar to the baseline values; SCH50911 completely prevented any significant
`decrease in frequency compared with baseline.
`
`17 (7)
`
`53.0 (19)
`
`—
`
`—
`
`Potential Treatment Strategies. Effects of potential
`treatment strategies on GHB-induced respiratory depression
`are given in Table 4. Administration of 50 mg/kg SCH50911
`5 min after GHB completely reversed the GHB-induced de-
`crease in respiratory rate, as shown in Fig. 4; there was no
`significant decrease in respiratory rate compared with base-
`line after the administration of SCH50911. In fact, a slight,
`but significant, increase in respiratory rate was observed at
`early time points in SCH50911-treated animals. Lower doses
`of 2.5, 5, and 10 mg/kg SCH50911 did not completely reverse
`GHB-induced respiratory depression, and significant de-
`creases in respiratory rate were still observed after antago-
`nist administration. Administration of 10 mg/kg SCH50911
`significantly improved all pharmacodynamic parameters,
`whereas 5 mg/kg improved only the ABEC and Emax and 2.5
`mg/kg had no significant effect on any pharmacodynamic
`parameter compared with that of GHB alone. Administration
`of 50 mg/kg SCH50911 also increased the nonrenal clearance
`
`230
`
`Morse et al.
`
`day precision and accuracy of the quality control samples
`are summarized in Table 1.
`Dose Dependence of GHB Pharmacokinetics/Phar-
`macodynamics. GHB administration in increasing intrave-
`nous doses displayed nonlinear pharmacokinetics, as shown
`in Table 2, similar to previous reports (Lettieri and Fung,
`1979; Morris et al., 2005). Renal clearance and the urinary
`excretion of GHB was almost negligible at the lowest dose of
`200 mg/kg but represented the predominant route of elimi-
`nation at the highest dose of 1500 mg/kg. The pharmacody-
`namic results of this experiment are shown in Fig. 1. Increas-
`ing doses of GHB resulted in a dose-dependent decrease in
`the parameter of respiratory rate, which was accompanied by
`a dose-dependent increase in tidal volume. Minute volume
`was unchanged with the 200 and 600 mg/kg doses but was
`significantly decreased with the 1500 mg/kg dose (95 ⫾ 18
`ml/min at baseline versus Emax of 54 ⫾ 24 ml/min; mean ⫾
`S.D., p ⬍ 0.05). Raw plethysmography traces displaying the
`change in respiratory pattern with GHB administration are
`shown in Fig. 2. As a result of this experiment, respiratory
`rate was considered the primary parameter of interest for
`assessment of receptors involved and potential treatment
`strategies. It was also determined in this experiment that
`1500 mg/kg GHB was the maximal dose that could be admin-
`istered without causing death; therefore, this dose was used
`for further investigation.
`Neurotransmitter Receptors Involved in GHB-In-
`duced Respiratory Depression. Effects of pretreatment
`with specific receptor antagonists are given in Table 3. Ad-
`ministration of the GABAB inhibitor, SCH50911 (150 mg/kg),
`before GHB, resulted in no significant decrease in respiratory
`rate nor a change in tidal volume compared with baseline, as
`displayed in Fig. 3. This inhibitor also increased the nonrenal
`clearance of GHB, but not the total clearance at this dose.
`Administration of the GABAA inhibitor, bicuculline methio-
`dide (5 mg/kg), before GHB, resulted in no change in the
`respiratory effects compared with those for GHB alone and
`had no significant effects on GHB pharmacokinetics.
`
`Page 5 of 10
`
`

`
`GHB-Induced Respiratory Depression
`
`231
`
`Downloaded from
`
`molpharm.aspetjournals.org
`
` at ASPET Journals on January 13, 2016
`
`Fig. 3. Effect of specific receptor in-
`hibitors on GHB-induced respiratory
`depression. GHB (1500 mg/kg) was ad-
`ministered intravenously, alone and af-
`ter pretreatment with the GABAB re-
`ceptor antagonist SCH50911 (150 mg/
`kg) and the GABAA receptor antagonist
`bicuculline methiodide (5 mg/kg). In-
`hibitors were administered intrave-
`nously 5 min before GHB. Data are pre-
`sented as mean ⫾ S.D.; n ⫽ 3 to 5.
`
`TABLE 4
`Effects of potential treatment strategies on the pharmacokinetics/pharmacodynamics of GHB (1500 mg/kg i.v.)
`Data are presented as mean (SD); n ⫽ 3 to 5. Control ⫽ administration of GHB 1500 mg/kg intravenously. SCH50911 and L-lactate were administered intravenously 5 min
`after GHB. L-Lactate was administered as a 66 mg/kg bolus followed by a 302.5 mg/kg/h infusion for 8 h. One-way analysis of variance followed by Tukey’s post hoc test was
`used to detect statistically significant differences in mean pharmacokinetic and pharmacodynamic parameters.
`
`Control
`
`SCH50911
`
`50 mg/kg
`
`10 mg/kg
`
`5 mg/kg
`
`2.5 mg/kg
`
`L-Lactate
`
`5 mg/kg SCH50911 ⫹
`L-Lactate
`
`Cl, ml 䡠 kg⫺1 䡠 min⫺1
`5.16 (0.70)
`ClR, ml䡠 kg⫺1 䡠 min⫺1
`3.18 (0.66)
`Clm, ml䡠 kg⫺1 䡠 min⫺1
`1.99 (0.17)
`10500 (2700)
`Frequency ABEC, breaths
`Frequency Emax, breaths/min
`17 (7)
`4.35 (1.3)
`Td, h
`a Significantly different from control (P ⬍ 0.05).
`b Significantly different from 5 mg/kg SCH50911 alone (P ⬍ 0.05).
`c Significantly different from L-lactate alone (P ⬍ 0.05).
`d—, no ABEC, Emax, orT d values could be calculated because respiration is similar to the baseline values; no significant decrease in frequency compared with baseline
`was observed after administration of 50 mg/kg SCH50911.
`
`6.17 (0.41)
`3.37 (0.038)
`2.80 (0.52)a
`—d
`—
`—
`
`6.13 (0.22)
`4.03 (0.21)
`2.09 (0.37)
`3690 (1440)a
`51 (3)a
`2.50 (0.20)a
`
`6.13 (0.23)
`3.78 (0.36)
`2.35 (0.17)
`5500 (1440)a
`44 (6)a
`3.15 (0.28)
`
`6.05 (0.69)
`3.56 (0.50)
`2.50 (0.19)
`8720 (513)
`33 (2)
`4.62 (1.2)
`
`6.40 (0.62)a
`4.22 (0.63)a
`2.19 (0.55)
`5470 (1550)a
`24 (5)
`2.45 (0.62)a
`
`7.61 (0.062)a,b,c
`5.28 (0.42)a,b
`2.33 (0.44)
`3170 (957)a
`45 (6)a
`2.17 (0.14)a
`
`Page 6 of 10
`
`

`
`232
`
`Morse et al.
`
`Downloaded from
`
`molpharm.aspetjournals.org
`
` at ASPET Journals on January 13, 2016
`
`Fig. 4. Effect of potential treatment
`strategies on respiratory rate after
`GHB administration. A, dose-depen-
`dent effects of SCH50911. B, effects of
`L-lactate and L-lactate and SCH50911
`combination therapy. All treatments
`were administered intravenously 5
`min after GHB (1500 mg/kg i.v.). Data
`are presented as mean ⫾ S.D.; n ⫽ 3
`to 5.
`
`of GHB, similar to the higher dose of this receptor antagonist,
`but this effect was not observed at lower doses. Administra-
`tion of the MCT inhibitor L-lactate significantly increased
`GHB renal and total clearance and resulted in significant
`decreases in the frequency ABEC and Td but did not improve
`the Emax. The combined administration of 5 mg/kg SCH50911
`plus L-lactate improved all pharmacodynamic endpoints com-
`pared with those for GHB alone. When L-lactate was admin-
`istered alone at the same dose as that given for GHB, the
`plasma lactate concentrations obtained with this dose were
`much lower in the absence of GHB, indicating an effect of
`GHB on lactate pharmacokinetics. A higher dose of 66 mg/kg
`plus 605 mg/kg/h L-lactate was then administered alone to
`achieve lactate concentrations similar to those obtained with
`GHB administration; ⬃1.5 mM mean increases in plasma
`lactate concentrations were obtained with this higher dose
`and with the lower dose administered concomitantly with
`GHB, as shown in Fig. 5. As displayed in Fig. 6, this higher
`
`dose of L-lactate had no significant effect on respiratory rate,
`tidal volume, or minute volume.
`
`Discussion
`Although abuse of GHB and its precursors has been recog-
`nized, no pharmacologic treatment for GHB overdose exists,
`and current treatment consists only of supportive care. Be-
`cause of the high rates of respiratory depression reported in
`both fatal and nonfatal cases of GHB overdose, this pharma-
`codynamic endpoint serves as a clinically relevant marker for
`the evaluation of potential GHB overdose treatment strate-
`gies. The current data indicate that the primary effect of
`GHB on respiration is a decrease in respiratory rate, accom-
`panied by a compensatory increase in tidal volume, allowing
`minute volume to remain constant until doses approach le-
`thality. This respiratory pattern is similar to that noted in
`some clinical cases of GHB overdose (Mason and Kerns,
`
`Page 7 of 10
`
`

`
`GHB-Induced Respiratory Depression
`
`233
`
`Fig. 5. Plasma lactate concentrations after administration
`of L-lactate alone and with GHB. Data are presented as
`mean ⫾ S.D.; n ⫽ 4 to 5. L-Lactate low-dose (LD) ⫽ 66
`mg/kg ⫹ 302.5 mg/kg/h. L-Lactate high-dose (HD) ⫽ 66
`mg/kg ⫹ 605 mg/kg/h. L-Lactate LD and HD were admin-
`istered alone at time 0. L-Lactate LD was administered 5
`min after GHB when administered concomitantly.
`
`Downloaded from
`
`molpharm.aspetjournals.org
`
` at ASPET Journals on January 13, 2016
`
`2002), indicating that our rat model is relevant for studying
`this endpoint. When the respiratory rate decreases substan-
`tially and tidal volume reaches a physiologic limit, minute
`volume decreases steeply with increases in GHB concentra-
`tions, resulting in fatality. Although we observed no fatalities
`at doses of 1500 mg/kg i.v. and lower, we observed fatality in
`approximately 50% of animals administered 1750 mg/kg,
`consistent with previous reports indicating that the LD50 of
`GHB administered intravenously in rats is 1700 mg/kg (La-
`borit, 1964). These results indicate that minute volume is an
`insensitive measure of GHB intoxication, because there is
`little change in this measure before death, and the primary
`parameter of interest in GHB-induced respiratory depression
`is respiratory rate.
`Our data indicate that the decrease in respiratory rate
`after GHB administration is mediated primarily by action at
`GABAB receptors, because of complete inhibition of the effect
`of GHB on this parameter with pretreatment of a GABAB
`receptor antagonist, SCH50911. This inhibitor also exhibited
`a surprising effect on GHB pharmacokinetics, significantly
`increasing its nonrenal clearance. This effect was probably
`not translated to increased total clearance because nonrenal
`clearance is a minor route of GHB elimination at 1500 mg/kg
`but may be responsible for some of the effects of this inhibitor
`observed on GHB toxicodynamics in studies at lower GHB
`doses. Although the primary mechanism of GHB-induced
`respiratory depression was evident from GABAB receptor

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket