throbber
Electroencephalographic effects and serum concentrations
`after intranasal and intravenous administration of
`diazepam to healthy volunteers
`
`Karsten Lindhardt,1 Sveinbjo¨ rn Gizurarson,2,3 Sigurjo´ n B. Stefa´nsson,4
`David R. O` lafsson2 & Erik Bechgaard1
`1The Royal Danish School of Pharmacy, Department of Pharmaceutics, Universitetsparken 2, 2100 Copenhagen, Denmark, 2Lyfjathroun hf,
`Hafnarbudir, Geirsgata 9, 101 Reykjavik, 3Department of Pharmaceutics, University of Iceland, 107 Reykjavik and 4Landspı´talinn,
`University Hospital of Iceland, Department of Neurology, 121 Reykjavik, Iceland
`
`Aims To evaluate the electroencephalographic (EEG) effects, blood concentrations,
`vehicle irritation and dose-effect relationships for diazepam administered nasally.
`Methods The study had a cross-over design with eight healthy volunteers (one drop
`out). It consisted of four legs with four different administrations: intranasal (i.n.)
`placebo, 4 mg diazepam i.n., 7 mg diazepam i.n. and 5 mg intravenous (i.v.) dia-
`zepam. Polyethylene glycol 300 (PEG300) was used as a vehicle in the nasal
`formulations to solubilize a clinically relevant dose of diazepam. Changes in N100,
`P200 and P300 brain event-related potentials (ERP) elicited by auditory stimulation
`and electroencephalographic b-activity were used to assess effects on neurological
`activity.
`Results The mean [95% confidence intervals] differences between before and
`after drug administration values of P300-N100 amplitude differences were x0.9
`[x6.5, 4.7], x6.4 [x10.1, x2,7], x8.6 [x11.4, x5.8] and x9.6 [x12.1, x7.1]
`for placebo, 4 mg i.n., 7 mg i.n. and 5 mg i.v. diazepam, respectively, indicating
`statistically significant drug induced effects. The bioavailabilities of 4 and 7 mg i.n.
`formulations, were found to be similar, 45% [32, 58] and 42% [22, 62], respectively.
`Conclusion The present study indicates that it is possible to deliver a clinically effec-
`tive nasal dose of diazepam for the acute treatment of epilepsy, using PEG300 as a
`solubilizer.
`
`Keywords: benzodiazepine, diazepam, EEG, electroencephalography, ERP, event-
`related potential, intranasal, nasal, PEG300, polyethylene glycol 300
`
`Introduction
`
`The present study was carried out to assess the intranasal
`administration of diazepam as a potential alternative to
`intravenous and rectal dosing in the treatment of acute
`epileptic seizures. A nasal spray is beneficial when a rapid
`onset of effect (within seconds or minutes) is required.
`Animal experiments have shown that
`the intranasal
`administration of diazepam may induce effects within
`5 min. In rabbits, a peak serum concentration is obtained
`about 5 min after
`the administration [1]. Diazepam
`
`Correspondence: Dr Sveinbjo¨ rn Gizurarson, University of Iceland, Department
`of Pharmaceutics, Hofsvallagata 53, 107 Reykjavik, Iceland. Tel.: 00354 5112020;
`E-mail: sg@lyf.is
`
`Received 16 October 2000, accepted 22 June 2001.
`
`has poor water solubility, but polyethylene glycol 300
`(PEG300), a vehicle causing relatively little local irritation,
`has been found to solubilize an expected clinically relevant
`dose (4–10 mg) of diazepam in the limited volume
`necessary for nasal administration [2].
`In an earlier clinical study a nasal dose of 2 mg diazepam
`was administered by use of PEG300 as the solubilizing
`vehicle [2]. Within 30 min the nasal bioavailability was
`found to be about 37%. The neurological measurements in
`this study were rather crude, and comprised parameters
`such as memory tests and the ability to catch a ruler. The
`quantification of drug effects on attention and vigilance
`was based on questionnaires. The results of this study
`showed only minor drug effects, probably because the dose
`was too low. Therefore,
`it was decided to administer
`higher nasal doses of diazepam (4 and 7 mg).
`
`f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 52, 521–527
`
`521
`
`AQUESTIVE EXHIBIT 1025 page 0001
`
`

`

`K. Lindhardt et al.
`
`The electroencephalographic (EEG) effects of benzo-
`diazepines are well known. Changes after drug adminis-
`tration have been observed in event-related potentials
`(ERP) and beta-activity [3–9]. The brain generates elec-
`trical waves of various wavelengths creating a spectrum,
`which may be divided into several
`frequency bands.
`The most important bands are found in the frequency
`range 8 –12 and 12–35 Hz, named the alpha and beta-
`activity, respectively. An increase of beta-activity has been
`found to be a more sensitive measure of benzodiazepine
`effect than a decrease in alpha-activity [10].
`Exposing study subjects to target tones (e.g. 2000 Hz
`auditory stimuli) among neutral tones (e.g. 1000 Hz audi-
`tory stimuli) generates ERPs. The neutral tones occur five
`times more frequently than the target. A positive wave
`appears 300 ms (P300) after the target tone. This wave is
`generated from the sensory discrimination of the target
`tone among the neutral tones. The P300 potential has
`been found to be particularly useful in measuring the
`intracerebral effects of benzodiazepines [8]. After both
`neutral and target tones a negative wave appears after
`100 ms (N100), but only limited changes appear after
`benzodiazepine administration [3]. As well as being related
`to the sedative and cognitive effects of benzodiazepines
`[8], changes in EEG, particularly the increase in beta-
`activity, has been found to correlate with anticonvulsant
`effect [10].
`Unrug et al. [3] found that the decrease in the P300
`potential after a 10 mg oral dose of diazepam was
`most pronounced at the vertex electrode. The peak of
`the P300 potential is usually identified as the most positive
`point in the waveform range between 200 and 400 ms and
`a change in the latency of this peak may also be useful in
`evaluating changes in P300 caused by diazepam [8].
`Fink et al. [9] found a linear correlation between the
`increase in EEG and beta-activity and blood concentrations
`of diazepam after oral administration to healthy volunteers.
`More recent studies of the pharmacological effects of
`benzodiazepines in man have been of nonblinded design,
`and only 3 out of 18 were controlled, emphasizing the need
`for additional well designed studies in this field [11].
`The aims of the present study were (1) to provide
`information on the pharmacodynamic response to nasally
`administered diazepam formulated in a polyethylene glycol
`300 vehicle (2) to evaluate and optimize EEG methods for
`measuring the neurological effects of diazepam, and (3) to
`define any relationship between the effect of diazepam on
`the EEG and serum concentrations of the drug.
`
`Methods
`
`Three male and five female healthy Caucasians, weighing
`84t17 kg, and between 20 and 40 years of age were
`studied (one of whom dropped out). They were asked not
`
`to drink alcohol during the entire study period and none
`was taking regular medication. Subjects received both
`written and oral information before giving their written
`consent. The National Icelandic Ethics Committee and
`the Icelandic Health Department approved the study.
`The study had a double-blind, randomized, cross-over
`design. Eight subjects received on separate occasions (1)
`placebo intranasal (i.n.) administration of PEG300, (2)
`4 mg diazepam i.n. solubilized in PEG300 (4 mg i.n.),
`(3) 7 mg diazepam i.n. solubilized in PEG 300 (7 mg i.n.),
`and (4) 5 mg diazepam intravenously (i.v.) administered in
`a commercially available formulation (Stesolid Novum1).
`The latter and PEG300 were obtained from Dumex-
`Alpharma A/S (Copenhagen, Denmark) and Union
`Carbide (Charleston, U.S.A.), respectively. In order to
`improve the spray properties of the viscous formulation, it
`was necessary to modify a unit-dose device from Pfeiffer
`(Radolfzell, Germany). Each device was filled to spray
`75 ml in each nostril (two devices per nasal round).
`
`Blood sampling
`Venous blood samples were taken at x10, x2, 3, 5, 8, 11,
`15, 20, 30, 45 and 60 min after drug administration. A
`commercially available enzyme-immunoassay (EIA) kit
`from STC Technologies, Inc. (Bethlehem, U.S.A.) was
`used for the analysis of diazepam in serum. The measure-
`ments were performed on a HTS7000 microplate reader
`from Perkin-Elmer (Wellelsly, U.S.A.) with u.v. detec-
`tion at 450 nm. Samples were centrifuged at 3200 g for
`10 min and serum was transferred to 1 ml cryotubes from
`NUNC (Copenhagen, Denmark) and stored at x80u C
`until analysis. Standards of 1, 2, 5 and 20 ng mlx1 (n=9)
`were analysed on separate days and a mean coefficient of
`variation was found to be 10% (range 7–13). The lowest
`level of detection was about 0.1 ng mlx1. All samples had
`concentrations higher than 1 ng mlx1.
`
`ERP and b-activity
`
`The hardware and software systems used for the EEG
`recordings and analyses were from Neuroscan1 (Sterling,
`U.S.A.). Nineteen silver scalp electrodes (F1, F2, F7, F3,
`Fz, F4, F8, T3, C3, Cz, C4, T4, T5, P3, Pz, P4, T6, O1,
`O2) were placed on the head according to the inter-
`national 10–20 system [12]. Electrodes placed on the
`left and right mastoid process (electrodes A1 and A2)
`were connected together and used as a reference. Two
`electrodes, one above the other below the left eye, were
`used to monitor eye movements. The impedance of the
`electrodes was tested before the recording started.
`The subjects sat in a chair during the recording and with
`their eyes shut. They were asked to listen to auditory
`stimuli delivered through earplugs to both ears. The neutral
`
`522
`
`f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 52, 521–527
`
`AQUESTIVE EXHIBIT 1025 page 0002
`
`

`

`stimulus was a 1000 Hz tone occurring five times more
`frequently than the 2000 Hz target tone. A preliminary
`test was performed to ensure the subjects were able to
`hear and separate the stimuli. The interstimulus-interval
`was 2 s, tone duration was 100 ms, and rise and fall
`time was 5 ms. The subjects were asked to tap with
`their finger and count when a target tone appeared. If
`subjects fell asleep, as happened a few times, especially
`in the i.v. group, they were gently awoken by touching
`their hands.
`The EEG was sampled at 200 Hz after low pass filtering
`at 40 Hz and high pass filtering at 0.3 Hz. Each
`auditory stimulus
`triggered a sampling of EEG that
`started 100 ms before the stimulus and had a total duration
`of 1280 ms
`(256 sampling points). The EEG epoch
`triggered by each auditory stimulus was stored for further
`analysis.
`The EEG was recorded before and after the subjects had
`received medication. Each recording session lasted about
`15 min during which 400–500 stimuli were delivered. The
`recordings were analysed by averaging the EEG epochs to
`the neutral and the target stimuli, respectively. Epochs
`were rejected when amplitudes exceeded t100 mV either
`in the lead across the eye or at the Fz, Cz or Pz electrodes.
`The peak of the P300 potential was defined by the highest
`potential between 200 and 500 ms and was located in the
`average ERP for each individual before and after treat-
`ment. The peak of the N100 potential was defined by the
`lowest potential between 50 and 200 ms. The Cz elec-
`trode resulted in the most sensitive effect measurements
`and was therefore chosen for the ERP calculations. To
`assess changes in beta activity, the mean amplitude and
`relative power spectrum were obtained after Fourier
`transformation of each epoch. The mean amplitude of
`the b-activity within different frequency bands between
`16 and 35 Hz was calculated.
`
`Diazepam intranasally administered
`
`Questionnaires
`
`The questionnaires were answered as soon as possible
`after each measurement. Subjects were asked to score a
`prefabricated list of various possible irritant effects from
`each formulation.
`
`Data analysis
`
`The area under the serum concentration-time curve from
`0 to 60 min (AUC(0,60 min)) was calculated using the
`trapezoidal method. AUC from 0 to 2 min for intravenous
`administrations were determined by extrapolation to zero
`by using logarithmic regression analysis on the initial two
`concentrations. A two-factor ANOVA was used to compare
`differences between pharmacodynamic measurements
`obtained by subtracting values after drug administration
`from pre-dose values. P300, P300-N100 differences and
`b-activities were tested. A two-sample t-test (one-sided)
`was used in the statistical analysis to compare the various
`data sets.
`
`Results
`
`Mean serum concentration-time profiles of seven subjects
`are shown in Figure 1. The mean bioavailability, Cmax and
`tmax, [95% confidence interval], for the 4 and 7 mg i.n.
`diazepam formulations were found to be; 45% [32, 58] and
`42% [22, 62], 99 ng mlx1 [83, 115] and 179 ng mlx1
`[126, 232], 18 min [11, 25] and 42 min [25, 59], respec-
`tively. tmax was significantly (P<0.05) higher after 7 mg
`i.n. administration than after 4 mg. The slower absorp-
`tion from the 7 mg dose was substantiated by differences
`in the AUCi.n./AUCi.v. ratio of the drug between the two
`formulations at the early time points (Table 1).
`A two-factor ANOVA was used to compare P300,
`P300-N100 amplitude differences and b-activity effects
`
`20
`
`40
`
`Time (min)
`
`60
`
`523
`
`AQUESTIVE EXHIBIT 1025 page 0003
`
`10000
`
`1000
`
`100
`
`10
`
`0
`
`Serum diazepam concentration (ng ml–1)
`
`Figure 1 Mean (ts.d.) serum
`concentration-time profiles after
`intranasal (i.n.) administration of 4 (#)
`and 7 mg (&) diazepam and 5 mg (m)
`diazepam intravenous (i.v.),
`respectively, to seven healthy subjects.
`
`f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 52, 521–527
`
`

`

`K. Lindhardt et al.
`
`Table 1 Mean AUCi.n./AUCi.v.ratios (expressed as percentage) at various times after the administration of 4 and 7 mg diazepam to seven healthy
`subjects.
`
`AUCi.n./AUCi.v. (%)
`Time (min)
`
`I.n.dose
`
`4 mg
`7 mg
`P valueb
`
`3
`
`12a
`7a
`0.07
`
`5
`
`14
`8
`0.05
`
`8
`
`17
`12
`0.09
`
`11
`
`21
`15
`0.14
`
`15
`
`25
`19
`0.17
`
`20
`
`29
`22
`0.12
`
`30
`
`34
`27
`0.09
`
`45
`
`40
`34
`0.19
`
`60
`
`45
`42
`0.63
`
`aOne AUC value was left out because it was thought to be an outlier, being more than three times the standard deviation above all the other values.
`bP values from the comparison between the two doses.
`
`Table 4 Mean (ts.d., n=7) EEG amplitude (nV) values of differences
`in the b-frequency range (16–35 Hz) at the vertex electrode between
`before and after drug administration.
`
`Formulation
`
`Placebo
`4 mg i.n.
`7 mg i.n.
`5 mg i.v.
`
`Difference between before
`and after administration
`
`95% confidenceb
`intervals
`
`64t163
`189t232*
`139t68*
`222t120*
`
`[x57, 184]
`[17, 360]
`[89, 189]
`[133, 342]
`
`aP<0.05(*), P<0.01(**), P<0.001(***) vs placebo.
`bNo significant differences were found between formulations or
`subjects.
`
`15
`
`10
`
`P200 P300
`
`300
`
`700
`
`1100
`
`Time (ms)
`
`Potential mV
`
`05
`
`–100
`–5
`
`–10
`
`–15
`
`N100
`
`Figure 2 Mean values from seven healthy subjects of the
`ERPs at the vertex electrode elicited by frequent nontarget
`events (1000 Hz tones, thin line) and rare target events
`(2000 Hz tones, thick line), respectively. Note the P300 evoked
`by the rare tones.
`
`The differences [95% confidence intervals] between the
`before and after values for the P300-N100 amplitude
`differences were x0.9 [x6.5, 4.7], x6.4 [x10.1, x2,7],
`x8.6 [x11.4, x5.8] and x9.6 [x12.1, x7.1] for
`placebo, 4 mg i.n., 7 mg i.n. and 5 mg i.v. diazepam,
`respectively. The overall means of the ERPs (all subjects)
`elicited, respectively, by neutral and target stimuli before
`treatment are shown in Figure 2 and those elicited by
`the target stimuli after treatment in Figure 3.
`
`Table 2 Mean (ts.d., n=7) in P300 potential (mV) at the vertex
`electrode between before and after administration.
`
`Formulation
`
`Placebo
`4 mg i.n.
`7 mg i.n.
`5 mg i.v.
`
`Difference between before
`and after drug administration
`
`x0.8t2.2
`x2.8t3.1
`x5.0t2.4*,a
`x5.6t2.8**
`
`95% confidenceb
`intervals
`
`[x3.0, 1.4]
`[x5.9, 0.3]
`[x6.7, x3.2]
`[x8.4, x2.7]
`
`aP<0.05(*), P<0.01(**), P<0.001(***) vs placebo.
`bNo significant differences were found between formulations or
`subjects.
`
`Table 3 Mean (ts.d., n=7) changes in P300-N100 potential
`differences (mV) at the vertex electrode between before and after drug
`administration.
`
`Formulation
`
`Placebo
`4 mg i.n.
`7 mg i.n.
`5 mg i.v.
`
`Difference between before
`and after drug administration
`
`x0.9t7.5
`x6.4t5.0*
`x8.6t3.8**
`x9.6t3.4***
`
`95% confidenceb
`intervals
`
`[x6.5, 4.7]
`[x10.1, x2.7]
`[x11.4, x5.8]
`[x12.1, x7.1]
`
`aP<0.05(*), P<0.01(**), P<0.001(***) vs placebo.
`bSignificant differences were found between formulations, but not
`between subjects.
`
`before and after treatment (Tables 2– 4). No significant
`difference was found between subjects. However for the
`P300-N100 amplitude differences a significant drug effect
`(P<0.05) was found. A significant reduction in P300
`amplitude was observed, using a two-sample t-test,
`compared with placebo, after 7 mg i.n. (P<0.05) and
`the 5 mg i.v. administrations (P<0.01), but not after the
`4 mg i.n. administration (Table 2). Significant decreases
`compared with placebo was also found in the P300-N100
`amplitude differences (P<0.05) (P<0.01) and (P<0.001)
`for the 4 mg i.n., 7 mg i.n. and 5 mg i.v. formulations,
`respectively (Table 3). The corresponding significance
`values for the beta-activity were (P<0.05) (P<0.05) and
`(P<0.05), respectively (Table 4).
`
`524
`
`f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 52, 521–527
`
`AQUESTIVE EXHIBIT 1025 page 0004
`
`

`

`Diazepam intranasally administered
`
`Mean differences [95% confidence intervals] on the
`P300-N100 measured between the placebo and drug
`treatment were x3.3 [x0.4, x6.1], x4.8 [x2.4, x7.2]
`and x5.8 [x3.9, x7.8]
`for
`the 4 and 7 mg i.n.
`formulations and the 5 mg i.v. formulation, respectively,
`all of which were statistically significant effect of diazepam
`was found for all
`formulations. The mean difference
`[95% confidence intervals] between 4 mg i.n and 7 mg
`i.n., 5 mg i.v., respectively, were x1.5 [x3.3, 0.3] and
`x2.6 [x4.1, x1.0],
`indicating statistical difference
`between 4 mg i.n. and 5 mg i.v. The mean difference
`[95% confidence] between 7 mg i.n. and 5 mg i.v. was
`x1.1 [x3.1, 1.0], indicating no statistical difference in
`the neurological effect between these two diazepam
`formulations.
`
`No shift in latency of the ERP components was found
`after diazepam administration, and therefore, the ERP data
`are based on changes in ERP amplitudes obtained at the
`vertex electrode.
`By averaging the ERP epochs evoked by target stimuli
`within each consecutive 2 min period (approximately 10
`epochs of rare stimuli), it was possible to determine how
`the P300-N100 difference changed with time for different
`formulations of the drug. In Figure 4, this is shown as
`change in the ratio relative to placebo of the P300-N100
`difference in drug treatment.
`The questionnaires revealed that the adverse effects
`following drug treatment were limited. Bitter taste after
`nasal administration, was the most frequently reported
`adverse event.
`
`300
`
`700
`
`1100
`
`Time (ms)
`
`0–2
`
`2–4
`
`4–6
`6–8
`Time intervals (min)
`
`8–10
`
`10–12
`
`048
`
`12
`
`–100
`–4
`
`–8
`
`–12
`
`Voltage (mV)
`
`1.0
`
`0.9
`
`0.8
`
`0.7
`
`0.6
`
`Ratio of P300-N100 relative to placebo
`
`Figure 3 Mean values from seven
`subjects of the ERPs at the vertex
`electrode after hearing a rare target
`event (2000 Hz tone) after
`administration of placebo (thick line),
`4 mg diazepam (thin line), 7 mg
`diazepam (dotted line) intranasally or
`5 mg diazepam (dashed line)
`intravenously. Note the negative peak
`at 100 ms (N100) and the positive
`peak at 300 ms (P300).
`
`Figure 4 Mean values from seven
`subjects of N100-P300 potential
`differences (obtained by averaging
`ERPs within each consecutive 2 min
`period) after administration of placebo
`intranasally, 4 mg diazepam intranasally
`(&), 7 mg diazepam intranasally (
`) or
`5 mg diazepam intravenously (
`).
`Values (range 0.6–1.0) are illustrated as
`ratios of the P300-N100 difference for
`drug relative to placebo.
`
`f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 52, 521–527
`
`525
`
`AQUESTIVE EXHIBIT 1025 page 0005
`
`

`

`K. Lindhardt et al.
`
`Discussion
`
`The present study indicates that it is possible provide a
`clinically effective nasal dose of diazepam using PEG300 as
`a solubilizer. This could provide an alternative route of
`administration in the treatment of epilepsy with benefits in
`acute situations.
`The most relevant components of the ERPs are the
`negative peak at 100 ms (N100), the positive peak at
`200 ms (P200) and the positive peak at 300 ms (P300),
`which are only observed after target stimuli. The data
`confirm previous suggestions that the P300-N100 ampli-
`tude possesses increased sensitivity to diazepam, relative to
`the use of P300 alone. This difference in sensitivity may be
`due to cancellation of base-line fluctuations, as both the
`negative and positive peaks would be equally affected by
`such fluctuations. The vertex electrode (Cz) was found to
`be the most sensitive scalp location for the evaluation of
`the effect of diazepam, which is consistent with literature
`reports [3, 9].
`The P300-N100 amplitude relative to placebo increased
`throughout
`the 12 min study period following i.v.
`diazepam, whereas the serum diazepam concentration
`shows a corresponding decrease to about one third of the
`initial concentration. This indicates that changes in the
`ERPs following diazepam administration do not directly
`reflect the blood concentration of the drug, as suggested
`previously [9]. A more likely explanation is that diazepam,
`which is a very lipophilic substance, progressively dis-
`tributes into the fatty tissue of the brain as the serum
`concentration falls, resulting in a delayed pharmaco-
`dynamic effect. The apparent rapid onset of effect from the
`nasal formulation can be explained by olfactory absorp-
`tion [13], where drug is delivered directly from the nasal
`cavity to the brain.
`A later drug absorption phase was evident for 7 mg i.n.,
`but not for the 4 mg formulation. This may be due to
`absorption from sites other than the nasal cavity,
`for
`example the buccal or the gastrointestinal tract. The rapid
`transfer (20 min) of drugs from the nasal cavity to the
`gastrointestinal tract may limit the absorption of dia-
`zepam, due to precipitation in the nose. This may be more
`pronounced for the 7 mg dose, explaining the higher
`initial clearance. As expected, the intravenous formula-
`tion results in very high initial drug concentration in the
`blood and can be considered as a positive control for the
`effects of diazepam on the EEG effect measurements.
`Adding a taste-adjusting agent in a possible future nasal
`formulation, for example orange, may reduce its bitter
`taste. However, this disadvantage may be insignificant
`compared with the benefit of the nasal formulation in view
`of the severity of the clinical indication.
`One subject was unwilling to continue in the study,
`partly due to nausea and vomiting occurring 4 –5 h after
`
`i.n. placebo administration. Intravenous administration
`made most subjects tired, which did not occur with the
`nasal formulations. For further evaluation of the nasal
`administration of diazepam to treat epilepsy, it may be
`appropriate to measure photosensitive epileptic responses
`because these may be considered as primary measures of
`anticonvulsant activity.
`The present study indicates that it is possible deliver a
`clinically effective nasal dose of diazepam using PEG300 as
`a solubilizer. The P300-N100 amplitude difference in the
`EEG was the preferred method for measuring the central
`nervous system effects of diazepam.
`
`We thank the head of the Department of Neurology, Landspı´talinn,
`University Hospital of Iceland Elias O´ lafsson. The study was
`supported by the Centre of Drug Delivery and Transport (a project
`grant from the Danish Medical Research Council) and by Nycomed
`Pharma A/S.
`
`References
`
`1 Bechgaard E, Gizurarson S, Hjortkær RK. Pharmacokinetic
`and pharmacodynamic response after intranasal administra-
`tion of diazepam to rabbits. J Pharm Pharmacol 1997;
`49: 747–750.
`2 Gizurarson S, Gudbrandson FK, Jo´ nsson H, Bechgaard E.
`Intranasal administration of diazepam aiming at the treatment
`of acute seizures: Clinical trial in healthy volunteers. Biol Pharm
`Bull 1998; 3: 385–394.
`3 Unrug A, van Luitelaar WCJM, Coles MGH, Coenen AML.
`Event-related potentials in a passive and active auditory
`condition: effects of diazepam and buspirone on slow wave
`positivity. Biol Psychol 1997; 46: 101–111.
`4 Engelhardt W, Friess K, Hartung E, Sold M, Dierks
`T. EEG and auditory evoked potential P300 compared
`with psychometric tests in assessing vigilance after
`benzodiazepine sedation and antagonism. Br J Anaesth
`1992; 69: 75–80.
`5 Pooviboonsuk P, Dalton JA, Curran HV, Lader MH. The
`effects of single dose of Lorazepam on event-related potentials
`and cognitive function. Human Psychopharmacol 1996;
`11: 241–252.
`6 Hayakawa T, Uchiyama M, Urata J, Enomoto T, Okubo J,
`Okawa M. Effects of small dose of triazolam on P300.
`Psychia Clin Neurosci 1999; 53: 185–187.
`7 Pang E, Fowler B. Discriminating the effects of triazolam
`on stimulus and response processing by means of
`reaction time and P300 latency. Psychopharmacology 1994;
`115: 509–515.
`8 Picton PW. The P300 Wave of the Human
`Event-Related Potential. J Clin Neurophys 1992;
`9: 456– 479.
`9 Fink M, Irwin P, Weinfeld RE, Schwartz MA,
`Conney AH. Blood levels and electroencephalographic
`effects of diazepam and bromazepam. Clin Pharmacol
`Ther 1976; 20: 184 –191.
`10 Mandema JW, Danhof M. Electroencephalogram effect
`measures and relationships between pharmacokinetics and
`
`526
`
`f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 52, 521–527
`
`AQUESTIVE EXHIBIT 1025 page 0006
`
`

`

`pharmacodynamics of centrally acting drugs. Clin Pharmacokin
`1992; 23: 191–215.
`11 Rey E, Tre´luyer JM, Pons G. Pharmacokinetic optimisation of
`benzodiazepine therapy for acute seizures. Clin Pharmacokin
`1999; 36: 409 – 424.
`
`12
`
`13
`
`Jasper H. The ten twenty electrode system of the International
`Federation. Electroencephalogr Clin Neurophysiol 1958;
`10: 371–375.
`Illum L. Transport of drugs from the nasal cavity to the
`central nervous system. Eur J Pharm Sci 2000; 11: 1–18.
`
`Diazepam intranasally administered
`
`f 2001 Blackwell Science Ltd Br J Clin Pharmacol, 52, 521–527
`
`527
`
`AQUESTIVE EXHIBIT 1025 page 0007
`
`

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