Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive...

12
EEG in non-clinical drug safety assessments: Current and emerging considerations Simon Authier a, , Marcus S. Delatte b , Mary-Jeanne Kallman c , Joanne Stevens d , Carrie Markgraf e a CiToxLAB North America, 445 Armand Frappier, Laval, QC H7V 4B3, Canada b Division of Anesthesia, Analgesia and Addiction Products (DAAAP), CDER, U.S. Food & Drug Administration, Silver Spring, MD, USA c Kallman Preclinical Consulting, Greeneld, IN 46140, USA d Department of Pharmacology, Merck Research Laboratories, West Point, PA 19486, USA e Safety Assessment, Merck Research Laboratories, Kenilworth, NJ 07033, USA abstract article info Article history: Received 10 February 2016 Received in revised form 4 March 2016 Accepted 7 March 2016 Available online 15 March 2016 Electroencephalogram (EEG) data in nonclinical species can play a critical role in the successful evaluation of a compound during drug development, particularly in the evaluation of seizure potential and for monitoring changes in sleep. Yet, while non-invasive electrocardiogram (ECG) monitoring is commonly included in preclin- ical safety studies, pre-dose or post-dose EEG assessments are not. Industry practices as they relate to preclinical seizure liability and sleep assessments are not well characterized and the extent of preclinical EEG testing varies between organizations. In the current paper, we discuss the various aspects of preclinical EEG to characterize drug-induced seizure risk and sleep disturbances, as well as describe the use of these data in a regulatory context. An overview of EEG technologyits correct application and its limitations, as well as best practices for setting up the animal models is presented. Sleep and seizure detection are discussed in detail. A regulatory perspective on the use of EEG data is provided and, tying together the previous topics is a discussion of the translational aspects of EEG. © 2016 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). Keywords: EEG Seizure Polysomnography Safety pharmacology Convulsion Neurotoxicity Sleep Spike Telemetry 1. Introduction A recent survey indicated that most drugs approved in Japan be- tween 1999 and 2013 with reported adverse drug reactions (ADRs) classied as seizures/convulsions in patients were not identied to have a seizure liability during preclinical development (Nagayama, 2015). When considering seizure/convulsion observed at any dose, only 25 out of 105 (23.8%) approved drugs showed concordance of pre- clinical and clinical data for seizurogenic effects based on ADRs. When observed in preclinical studies, seizures/convulsions were identied in repeat toxicology studies (64%), proconvulsion safety pharmacology studies (40%) or in other safety pharmacology studies (28%). Proconvulsion safety pharmacology studies typically include models aimed to characterize the risk of drug-induced seizures such as EEG studies to monitor for ictal activity and seizure threshold tests. Other safety pharmacology studies include a wide range of pharmacology models (e.g. cardiovascular, respiratory, gastrointestinal and even other neurological models) which are dened under the ICH S7A guide- line (U.S. Food and Drug Administration, 2001). Industry practices as they relate to preclinical seizure liability assessments are not well char- acterized and the extent of preclinical seizure liability testing varies be- tween organizations (Authier et al., 2016). Spontaneous seizures are reported in various species including rats (Nunn & Macpherson, 1995; Satomoto et al., 2012) and dogs (Bielfelt, Redman, & McClellan, 1971) and it is crucial to differential spontaneous seizures from drug- induced ictal activity. Susceptibility to drug-induced seizures differs be- tween species (Bassett et al., 2014) but also between age groups (Himmel, 2008) within the same species rendering translation of pre- clinical results to humans challenging. Irrespective of the limitations when using animal models in drug development, preclinical seizure lia- bility testing strategies aim to succeed at risk identication and support clinical trial risk management. In a recent survey on preclinical neurotoxicology investigations, a minority of participants reported using pre-dose electroencephalog- raphy (EEG) (Authier et al., 2016) to conrm suitability of the animals for inclusion on study. As technology advances have in- creased the availability of non-invasive EEG monitoring and analysis (Pouliot et al., 2015), typical safety testing paradigms may need to be challenged. Tremors and other behavioral effects such as ataxia, myoclonus or emesis are often observed in early toxicology investigations such as Journal of Pharmacological and Toxicological Methods 81 (2016) 274285 Corresponding author. E-mail address: [email protected] (S. Authier). http://dx.doi.org/10.1016/j.vascn.2016.03.002 1056-8719/© 2016 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). Contents lists available at ScienceDirect Journal of Pharmacological and Toxicological Methods journal homepage: www.elsevier.com/locate/jpharmtox

Transcript of Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive...

Page 1: Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive electrocardiogram (ECG)monitoringiscommonly included inpreclin-icalsafety studies,pre-doseorpost-doseEEGassessments

Journal of Pharmacological and Toxicological Methods 81 (2016) 274–285

Contents lists available at ScienceDirect

Journal of Pharmacological and Toxicological Methods

j ourna l homepage: www.e lsev ie r .com/ locate / jpharmtox

EEG in non-clinical drug safety assessments: Current andemerging considerations

Simon Authier a,⁎, Marcus S. Delatte b, Mary-Jeanne Kallman c, Joanne Stevens d, Carrie Markgraf e

a CiToxLAB North America, 445 Armand Frappier, Laval, QC H7V 4B3, Canadab Division of Anesthesia, Analgesia and Addiction Products (DAAAP), CDER, U.S. Food & Drug Administration, Silver Spring, MD, USAc Kallman Preclinical Consulting, Greenfield, IN 46140, USAd Department of Pharmacology, Merck Research Laboratories, West Point, PA 19486, USAe Safety Assessment, Merck Research Laboratories, Kenilworth, NJ 07033, USA

⁎ Corresponding author.E-mail address: [email protected] (S. Authier)

http://dx.doi.org/10.1016/j.vascn.2016.03.0021056-8719/© 2016 The Authors. Published by Elsevier Inc

a b s t r a c t

a r t i c l e i n f o

Article history:Received 10 February 2016Received in revised form 4 March 2016Accepted 7 March 2016Available online 15 March 2016

Electroencephalogram (EEG) data in nonclinical species can play a critical role in the successful evaluation of acompound during drug development, particularly in the evaluation of seizure potential and for monitoringchanges in sleep. Yet, while non-invasive electrocardiogram (ECG)monitoring is commonly included in preclin-ical safety studies, pre-dose or post-dose EEG assessments are not. Industry practices as they relate to preclinicalseizure liability and sleep assessments are not well characterized and the extent of preclinical EEG testing variesbetween organizations. In the current paper, we discuss the various aspects of preclinical EEG to characterizedrug-induced seizure risk and sleep disturbances, aswell as describe the use of these data in a regulatory context.An overview of EEG technology—its correct application and its limitations, as well as best practices for setting upthe animal models is presented. Sleep and seizure detection are discussed in detail. A regulatory perspective onthe use of EEG data is provided and, tying together the previous topics is a discussion of the translational aspectsof EEG.

© 2016 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license(http://creativecommons.org/licenses/by/4.0/).

Keywords:EEGSeizurePolysomnographySafety pharmacologyConvulsionNeurotoxicitySleepSpikeTelemetry

1. Introduction

A recent survey indicated that most drugs approved in Japan be-tween 1999 and 2013 with reported adverse drug reactions (ADRs)classified as seizures/convulsions in patients were not identified tohave a seizure liability during preclinical development (Nagayama,2015). When considering seizure/convulsion observed at any dose,only 25 out of 105 (23.8%) approved drugs showed concordance of pre-clinical and clinical data for seizurogenic effects based on ADRs. Whenobserved in preclinical studies, seizures/convulsions were identified inrepeat toxicology studies (64%), proconvulsion safety pharmacologystudies (40%) or in other safety pharmacology studies (28%).Proconvulsion safety pharmacology studies typically include modelsaimed to characterize the risk of drug-induced seizures such as EEGstudies to monitor for ictal activity and seizure threshold tests. Othersafety pharmacology studies include a wide range of pharmacologymodels (e.g. cardiovascular, respiratory, gastrointestinal and evenother neurological models)which are defined under the ICH S7A guide-line (U.S. Food and Drug Administration, 2001). Industry practices as

.

. This is an open access article under

they relate to preclinical seizure liability assessments are not well char-acterized and the extent of preclinical seizure liability testing varies be-tween organizations (Authier et al., 2016). Spontaneous seizures arereported in various species including rats (Nunn & Macpherson, 1995;Satomoto et al., 2012) and dogs (Bielfelt, Redman, & McClellan, 1971)and it is crucial to differential spontaneous seizures from drug-induced ictal activity. Susceptibility to drug-induced seizures differs be-tween species (Bassett et al., 2014) but also between age groups(Himmel, 2008) within the same species rendering translation of pre-clinical results to humans challenging. Irrespective of the limitationswhen using animal models in drug development, preclinical seizure lia-bility testing strategies aim to succeed at risk identification and supportclinical trial risk management.

In a recent survey on preclinical neurotoxicology investigations, aminority of participants reported using pre-dose electroencephalog-raphy (EEG) (Authier et al., 2016) to confirm suitability of theanimals for inclusion on study. As technology advances have in-creased the availability of non-invasive EEG monitoring and analysis(Pouliot et al., 2015), typical safety testing paradigmsmay need to bechallenged.

Tremors and other behavioral effects such as ataxia, myoclonus oremesis are often observed in early toxicology investigations such as

the CC BY license (http://creativecommons.org/licenses/by/4.0/).

Page 2: Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive electrocardiogram (ECG)monitoringiscommonly included inpreclin-icalsafety studies,pre-doseorpost-doseEEGassessments

275S. Authier et al. / Journal of Pharmacological and Toxicological Methods 81 (2016) 274–285

maximum tolerated dose (MTD) studies. MTD studies are conductedduring drug development as part of the toxicology investigations as de-fined under the ICH guideline M3(R2) (U.S. Food and DrugAdministration, 2010). Once the MTD is identified, the drug dose levelsthat induce significant adverse effects may never be used again in theorganized sequence of preclinical drug safety testing studies. A commonconcern when tremors are present is the presence of underlying abnor-mal EEG activity. Surface ECGmonitoring is commonly included in pre-clinical toxicology studies but EEG assessments are classicallyintroduced only once a neurological concern is identified. MonitoringEEG duringMTDor repeat dose toxicology studiesmay represent an op-portunity for early identification of a CNS risk. With older patient popu-lations recognized to have an increased seizure incidence (Vélez &Selwa, 2003), this concern may be of increased clinical relevance giventhe life-threatening consequences of status epilepticus. Beyondseizurogenic risks, a number of drugs in development may alter sleeparchitecture (Rachalski et al., 2014) with potential negative impactson the patient population. Selective serotonin reuptake inhibitors(SSRIs) are widely prescribed but are also associated with sleep distur-bance (Ferguson, 2001) such as delayed REM sleep onset, increaseawakenings and reduce REM sleep. Here we discuss the various aspectsof preclinical EEG assessments to characterize seizure risk and also in-vestigate potential drug-induced sleep disturbances.

2. Fundamentals of EEG

To fully appreciate the role of EEG in nonclinical safety evaluation, anunderstanding of the fundamentals of the technology is important. Thefundamentals of EEG will detail what underlies the generation of EEGwaves, both from an anatomical and an instrumentation perspectiveand will review descriptive versus interpretation of EEG waveform pat-ters as well as describing typical normal EEG patterns.

2.1. What is EEG?

EEG is the recording of electrical activity from the brain's corticalsurface. Neuronal output is in μV, unlike the mV electrical signals fromrecording an ECG, and needs to be amplified by 106 to be displayed.Most of the EEG's electrical signal arises from neuronal post-synapticpotentials (PSP). Action potentials are too small and too short to record.PSPs can be excitatory (EPSP), causing the post-synaptic neuron to fire,or can be inhibitory (IPSP), causing the post-synaptic neuron not to fire.The combination of EPSPs and IPSPs induce current flow around neu-rons, which is recorded as EEG. The complex neuronal activity frommil-lions of cortical neurons generates the irregular EEG signal thattranslates into seemingly random and changing waveforms (Fig. 1). Bycontrast an evoked potential is an integrated signal that is synchronizedby a precipitating stimulus such as a noise or flashing light.

2.2. EEG instrumentation

While a wide range of EEG electrode types can be used(Galanopoulou et al., 2013), the most common use for nonclinical EEGis from the cortical surface. This recording can be accomplished usingscalp electrodes in a restrained subject, or by using telemetry, consistingof surgically implanted electrodes that send signals to a remote receiver.

Fig. 1.A single channel of EEG, showing the richmix of frequencies and amplitudes that compristo-second mix of amplitudes and frequencies, but also shows larger rhythmic oscillations charoscillations is partially due to the interaction between the thalamus and cortex. It is also based

Specialized applications may use depth electrodes surgically implantedinto the parenchyma of the brain (frequently into the hippocampus orthalamus). However obtained, the signal is amplified, filtered, displayedand recorded for analysis. Human EEG uses a system of standard place-ment of scalp electrodes, the 10–20 system (Jasper, 1958). Digital re-cording from this array allows the data to be displayed in differentmontages, which helps in defining abnormal waveforms and in localiz-ing the source of the abnormality. EEG in nonclinical species such as ro-dents, a standard electrode placement is not essential while a standardplacement is typically beneficial in larger species (e.g. non-humanprimates).

2.3. Interpretation of EEG patterns

For clinical and nonclinical applications, reading and understandingEEG waveforms is based on a systematic and organized process to rec-ognize abnormal from normal patterns. Interpretation of a typical10 second strip of 3-channel EEG from a non-human primate (Fig. 2)will require an exhaustive investigation of the context in which this ac-tivity was recorded (Table 1). For pre-seizure detection, the typical pat-tern is the spike. Spike morphology is generally electro-negative(deflects up first), the rise is faster than the fall, it is paroxysmal, is20–80 msec in duration and is of high voltage: 200–300 μV (Fig. 3). Aprecise description is essential when identifying an EEG pattern as nor-mal or pathological. For example, a 3 Hz spike and wave pattern is clas-sic finding in absence seizure (Panayiotopoulos, 1999); 2 Hz spike andwave typical for a seizure disorder while 6 Hz spike and wave is a nor-mal EEG variant identified as “14 and 6 positive spikes” or “ctenoids”(Bassett et al., 2014; Nidermeyer & Croft, 1961).

A number of normal EEG variants can be mistaken for seizures.Wickets (Fig. 4) are sharply contoured waves with a rhythmic frequen-cy at 7–11Hz thatwerefirst described by Reiher and Lebel (1977). Theyresemble theGreek letter “mu” and are often seen in drowsiness or lightsleep. Wickets may be misdiagnozed as epilepsy (Krauss, Abdallah,Lesser, Thompson, &Niedermeyer, 2005). Other commonEEGmorphol-ogies mimicking epileptiform discharges include hyperventilation-induced slowing, phantom spike-and-wave, hypnagogic and hypno-pompic hypersynchrony (Azzam & Bhatt, 2014; Benbadis & Tatum,2003). Increased synchrony (Fig. 5) is common during sleep stage tran-sitions and hypnagogic and hypnopompic hypersynchrony are consid-ered normal variants of drowsiness that may be misdiagnozed asseizure activity. The morphology of rhythmic mid-temporal discharges(RMTD; previously called psychomotor variant) shows patterns thatare notched and flat-topped, lasting 1–10 s (Fig. 6).

Artifacts are also a major consideration during EEG interpretation. Itis important to distinguish patterns generated from the brain from arti-facts created by factors outside the CNS. Movement is a frequently seenartifact, as muscles generate larger voltage signals than do neurons.Movement artifact is not only from whole body movement but can becaused by tongue or eye movements. Tongue movements cause thebaseline to undulate. Use of an ocular electrode placed above the eyecan help detect and localize eye movements. Usually, movement arti-facts affect scalp electrodes more often than implanted telemetry sys-tem electrodes. One exception in nonclinical species is chewing:chewing, particularly in monkeys or dogs (Fig. 7A and B), is frequentlyseen with EEG telemetry as the animals are free to move around the

es the normal EEG, in this example, from a Beagle dog. EEG consists not only of the second-acterized by slower frequencies. Physiologically, the underlying mechanism of these largeon the intrinsic rhythmic capacity of the large neuronal networks in the cortex.

Page 3: Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive electrocardiogram (ECG)monitoringiscommonly included inpreclin-icalsafety studies,pre-doseorpost-doseEEGassessments

Fig. 2. EEG from surface electrodes in a non-human primate in the high dose group at 2 hpost-dose; Tmax for this drug is 4 h and the drug is CNS-active.

Fig. 3. Typical spike morphology identified at EEG.

276 S. Authier et al. / Journal of Pharmacological and Toxicological Methods 81 (2016) 274–285

cage and engage in typical daily activities. Chewing activity can be dif-ferentiated from EEG by the presence of high frequency and high ampli-tude electromyographic (EMG) transients with typical crescendo/decrescendo profiles owing to muscular contractions. EMG is not ex-haustively discussed in the current publication but may be used to as-sess convulsion, activity level or sleep stages. A wide range of musclegroups can be monitored in laboratory animals including the temporalmuscles, neckmuscles or limbs. Theuse of temporalmuscles can beuse-ful in the interpretation of EEG traces from an electrode montage usingthe 10–20 system as the EMG activity can be compared to thematchingEEG traces to confirm artifacts. Neck muscles are typically used in sleepstudies (Rachalski et al., 2014). Videomonitoring can also be consideredfor interpretation of EEG (Authier et al., 2009). Video-EEG is consideredthe gold standard in clinical neurology and similar methodologies canbe applied to safety pharmacology studies.

3. EEG analysis

3.1. qEEG and spectral analysis

Spectral analysis is recognized as a common clinical (Fisher,Scharfman, & deCurtis, 2014) and preclinical (Bassett et al., 2014) toolfor data analysis for sleep and seizure assessments. EEG signals was ad-vanced in the 1970s with the advent of qEEG (or quantitative EEG), giv-ing added clarity and information of EEG signals by analyzing itspatterns using computer and mathematical methods. Fast FourierTransform (FFT) can be used to quantify the EEG power across a rangeof frequencies (Sterman, 1981). FFT converts the EEG signal to a repre-sentation of the power (cumulative amplitude) across the range of fre-quencies (Fig. 8A–D). An increase in higher frequencies can be observedprior to seizurewith various positive control drugs (Engel, Bragin, Staba,& Mody, 2009; Zijlmans, Jacobs, Zelmann, Dubeau, & Gotman, 2009).Conversely, an increase in qEEG powers may also be observed withdrugs that are not seizurogenic such as diazepam (Authier et al.,2014), highlighting the diversity of qEEG profiles which may beobserved.

qEEG changes may differ between brain regions adding to the inter-pretation considerations. The use of frontal, parietal and/or occipitalEEG derivation(s) needs to be weighted in the selection of the preclini-cal EEGmodel given the limitations generally caused by hardware spec-ifications but also cranial anatomy in some species. EEG derivations that

Table 1Generic questions to address during EEG interpretation.

Is the pattern present during drug treatment and/or baseline?If during treatment only, when does it occur post-dose?How does this timing relate to the pharmacokinetics of the drug?Are there any concommitant behavioral abnormalities?What is the frequency (Hz) and amplitude (μV) of the pattern?What is the morphology of the wave in comparison to surrounding traces?Does it occur over the top of normal rhythm or does it disrupt normal rhythm?Does it occur in complexes or singly?Is the pattern continuous or transient?Does the pattern recur?

are located in the midline sagittal plane are often preferred to conductqEEG investigations in all laboratory species due to the limited cranialmuscles present at this level therefore minimizing the interferencescaused by electromyographic (EMG) activity.

Social interactions and circadian cycle are key considerations in thestudy design of qEEG studies. Comparison of time-matched qEEG datafrom control and treated periods from the same animal can be used toevaluate potential drug effects. Interactions with congeners are highlybeneficial to the animals butwill also alter qEEGparameters. In this con-text, a drug inducing significant effects on the behavior of an animal inthe room may affect the qEEG profile of all animals in the same roomas communication between individuals occurs. To avoid bias causedby pharmacologically modified behavioral interactions, a qEEG studydesign would typically involve dosing on different days or in differentrooms on the same day.

Like most physiological parameters, qEEG is highly influenced by thecircadian cycle and proper interpretation usually relies on comparisonof data captured precisely at the same time of the day in the same animals(Authier et al., 2012). Owing to circadian cycle effects, a qEEG cross-overdesignwill often control the dosing timebut husbandry and feeding activ-ities should also be completed precisely at the same time of the day for alltreatment sessions. As for most other telemetry studies, the presence ofthe technical personnel in the room should be avoided when planningqEEG analysis. In large animal species, maintaining the same technicalpersonnel throughout the study will also increase data stability giventhe potential stress associated with a new caretaker. Despite a widerange of factors that need to be controlled inpreclinical qEEG, spectral anal-ysis remains one of themost sensitive endpoint with direct applicability insafety pharmacology studies and safety assessments with the ability toidentify low amplitude effects with a limited number of individuals.

3.2. Automated or manual EEG analysis in the assessment of seizurogeniceffects

Software for automated seizure detection on pre-clinical EEG tracesare typically designed to identify spikes trains (Authier et al., 2009)while epilepsy detection algorithms available for patient EEG allow fordetection of ictal and interictal traces (Acharya, Sree, Swapna, Martis,

Fig. 4. Example of human EEG showing wickets in the first and third traces.

Page 4: Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive electrocardiogram (ECG)monitoringiscommonly included inpreclin-icalsafety studies,pre-doseorpost-doseEEGassessments

Fig. 5. Increased synchrony in EEG from a freely moving cynomolgus monkey using implanted telemetry.

277S. Authier et al. / Journal of Pharmacological and Toxicological Methods 81 (2016) 274–285

& Suri, 2013). Normal and ictal EEG morphologies are species and indi-vidual specific and also vary depending on the electrode montage used.To account for these variations, it is generally useful to adapt the detec-tion algorithm to the experimental conditions that are used.While spiketrain detection is an important aspect of EEG analysis, a plethora ofother changes may be indicative of a seizure risk and manual reviewof EEG traces by an electroencephalographer is usually an importantpart of the analysis. Abnormal EEG activitymay occurwith concomitantclinical signs and systematic review of EEG traces at times when clinicalsigns were noted is often appropriate. For seizure activity mediated vianeuronal receptors or other concentration dependent mechanisms, theabnormal EEG activity may exhibit an incidence distribution correlatedwith higher plasma and CNS tissue exposure. Pre-defined manual re-view of EEG traces distributed over the monitoring period based onthe expected pharmacokinetics (PK) of the compound can be relevantand increase sensitivity to detect abnormal EEG activity or biomarkersof increased seizure risk.

4. Non-clinical sleep assessments

Preclinical sleep assessment is gaining rising importance in the ad-vancement of a pharmaceutical candidate as drug companies recognizethe value of translatable biomarkers that not only are predictive of effi-cacy but can also demonstrate engagement of its intended target aswellas detect adverse events early on in the drug discovery process. In addi-tion, the evaluation of sleep has been used as an endpoint to discovernovel therapies for insomnia (Winrow et al., 2011) and epilepsy aswell as a diagnostic for sleep disorders. Assessing sleep early on indrug discovery has become important not only when considering theeconomic burden of sleep loss ($100 billion US; Stoller, 1994) butwith the aging population, polypharmacy is more common and sleepdisruptions due to a new medication would almost certainly decreasecompliance with long term treatment. Sleep architecture is well studiedin all species, including humans, and many of its features are

Fig. 6. Example of human EEG showing rhythmic mid-temporal discharges (RMTD)(R:right side, L: Left side).

remarkably similar frommouse toman,making EEG one of only a hand-ful of highly translatable central nervous system biomarkers (Ivarsson,2009; Paterson, Nutt, & Wilson, 2011; Veasey et al., 2000).

The assessment of sleep stages is referred to as polysomnography,which requires the electrophysiological collection of up to three coremeasures. EEG measures the spontaneous electrical activity over singleor multiple brain regions while electromyogram (EMG)measures mus-cular electrical activity, and electrooculogram(EOG) assesses eyemove-ments. Biopotential activity of these core measures can be detected inanimals with surgically implanted electrodes implanted over the cortex(known as ECoG or electrocorticogram) for chronic sleep stage assess-ments and for frequency changes (see below). This method offers 24-hr/7-day EEG data collection, including temperature, locomotor activityandheart rate, all while the animal is free to roam in its home cagewith-out any tethering limiting movement. The combination of these mea-sures is used to define individual sleep stages during individualanalysis epochs, usually not greater than 30 s. A trained human EEGscorer or validated computer algorithm assigns a score of the predomi-nant sleep pattern within each epoch, using gold standard scoring rulesestablished by Rechshaffen & Kales in 1968 (Rechtschaffen & Kales,1968) and recently updated by the American Academy of Sleep Medi-cine (AASM) in 2007 (Iber, Ancoli-Israel, Chesson, & Quan, 2007). Theoverall sleep architecture can then be determined from the collectionof individual scoring epochs over the duration of the recording or duringa period of interest.

Wake patterns are easily recognized by the trained eye by the pres-ence of desynchronized, high frequency, low amplitude EEG, increasedmuscle tone and rapid eye movements. Sleep patterns are brokendown into three distinct non-REM stages (termed N1, N2 and N3) andREM sleep (rapid eye movement), each marked with distinct featuresthat help define them. N1 is a transitionary stage into falling asleep,marked by low voltage EEG and the beginning of slow eye movements.N2 has moderate voltage EEG with the introduction of sleep spindlesand K-complexes. As the individual progresses into N3, increased syn-chrony occurs in EEG as delta waves appear, voltage is high and assuch this stage is defined as “slow wave or delta sleep”. REM sleep ischaracterized by EEG that is desynchronized (as in wake butwith a pre-dominance of theta power) while muscle tonemeasured by EMG regis-ters atonia, and EOG measures the appearance of distinct bursts ofcharacteristic eye movements (Iber et al., 2007). Consolidated, diurnalsleepers such as non-human primates and humans, normally cyclethroughout the various non-REM stages during their sleep phase beforeultimately reaching REM sleep. EEG/EMG/activity are assessed in poly-phasic rodents (transition rapidly through sleep stages) typically byassigning sleep stages to either active wake, quiet sleep, deep sleep orREM sleep (Toth & Bhargava, 2013). EOG activity is typically not

Page 5: Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive electrocardiogram (ECG)monitoringiscommonly included inpreclin-icalsafety studies,pre-doseorpost-doseEEGassessments

Fig. 7. A. Chewing artifact at EEG recorded with telemetry in a freely moving jacketed Beagle dog B. Chewing artifact at EEG recorded with telemetry in a freely moving cynomolgusmonkey.

278 S. Authier et al. / Journal of Pharmacological and Toxicological Methods 81 (2016) 274–285

assessed in rodents, as these are difficult electrodes to implant andmaintain long term in this species.

qEEG has been shown to be a powerful and dynamic tool, as bursts(or power) of sinusoidal waves have been linked to certain mentalstates such as sedation or stimulation and have even been validated asa biomarker for disorders such as ADHD (increased theta-beta powerratio) (Leiser, Dunlop, Bowlby, & Devilbiss, 2011). Recent innovativedrug discovery approaches have compared frequency changes relativeto vehicle within certain sleep stages, allowing for better comparisonsto standard of care treatments for sleep disorders such as insomnia(Fox et al., 2013). EEGpatterns consist offive basic frequencies, alternat-ing between states of increased and decreased synchrony within thesignal, dependent on the sleep pattern the animal is in at the time ofmeasurement. At its slowest frequency and most synchronized state,delta activity is detected and ranges between 0.5–4 Hz and is associatedwith deep sleep. With increased frequency and desynchrony, delta isterminated and transitions into theta activity (4–8 Hz). Theta activityis often associated with REM sleep. Alpha activity (8–12 Hz) is readilyseen in relaxed or quiet wakefulness. Sigma activity (12–16 Hz) is typ-ically present in the awake state. Frequencies greater than 16 Hz are re-ferred to as beta (16–30 Hz) and gamma (N30 Hz) and are detectedduring active mental activity (Jobert et al., 2012). As a general rule,

Fig. 8. A. Fast Fourier Transform (FFT) on EEG (Cz–Oz) from a telemetered cynomolgus monkey200 mV). B. Fast Fourier Transform (FFT) on EEG (Cz–Oz) from a cynomolgus monkey duringfrom 0 to 70 mV). C. Fast Fourier Transform (FFT) on EEG (Cz–Oz) from a cynomolgus monkeaxis from 0 to 70 mV). D. Fast Fourier Transform (FFT) on EEG (Cz–Oz) from a cynomolgus mfrom 0 to 70 mV).

the cumulative absolute amplitude as calculated by FFT at lower fre-quencies (e.g. delta) will be lower than values for higher frequencies(e.g. beta or gamma). Hence, pharmacological evaluations typicallyrely on relative changes within a given power band (Bassett et al.,2014).

Typical study designs for the assessment of sleep architecture/fre-quency changes in animals involve evaluating the core measures(qEEG only requires EEG) asmentioned above with the potential exclu-sion of periods with high EMG activities. Chronic dosing studies prefer-ably involve pre-screening animals in order to power studies withanimals that have high quality EEG/EMG/EOG signals, good baseline cir-cadian patterns and are void of paroxysmal EEG activity. After baselineactivity has been assessed, animals are assigned to a chronic, multipleday dosing regimenwith either a parallel or cross-over design (random-ly assigned to drug first or vehicle first treatment and then crossed overto the opposite regime). Cross-over studies are preferable, as each ani-mal can act as their own control when a manipulation (drug or someother treatment) is compared to a negative control (either vehicle orsham). The use of genetically manipulated mice and rats or knockoutscan add value to pharmacological experiments as they can demonstratethe dependence of the drug's effects on a given target or gene product inanimals with a targeted genetic disruption. Knockouts serve as their

during generalized seizurewith a peak amplitude at 6 Hz (Graph amplitude axis from 0 tothe post-ictal period with significant attenuation of all frequencies (Graph amplitude axisy during deep sleep (stage N3) with an increase in lower frequencies (Graph amplitudeonkey during active wake with an increase in higher frequencies (Graph amplitude axis

Page 6: Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive electrocardiogram (ECG)monitoringiscommonly included inpreclin-icalsafety studies,pre-doseorpost-doseEEGassessments

279S. Authier et al. / Journal of Pharmacological and Toxicological Methods 81 (2016) 274–285

Page 7: Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive electrocardiogram (ECG)monitoringiscommonly included inpreclin-icalsafety studies,pre-doseorpost-doseEEGassessments

280 S. Authier et al. / Journal of Pharmacological and Toxicological Methods 81 (2016) 274–285

own controls and comparisons can be made against wild-typesindividuals.

Other than drug exposure, several factors can impact sleep architec-ture. Factors such as increased stress, illness, and changes in social/physical environment(s) have been shown to be disruptive to normalphysiological circadian patterns (Bruse et al., 2012; Irvine, White, &Chan, 1997). EEG studies in animals should aim to keep extraneous, un-controlled factors at a minimum. Normal and uncontrollable changesduring sleep include changes in respiratory drive which may inducechanges to sleep patterns. With the increased incidence of diagnosis ofsleep apnea and an aging population enduring polypharmacy, sleepstudies undertaken early in drug development can protect patientsdown the line who are already at risk for sleep issues. EEG studiesdone preclinically can also inform on efficacy, qualify normal EEG activ-ity and identify potential adverse events of a drug (and ultimately affectprescription rate down the line), as well as outline risks associated withsleep loss and changes in sleep, making it an extremely powerful tool toassess brain activity. While some methodological & analytical differ-ences do exist when assessing EEG in animals vs. humans, the sleep/wake systems in the brain are remarkably similar, allowing for translat-able, pharmacodynamic assessment of pharmaceutical candidates earlyon in the drug discovery process.

5. Species differences and interpretation of EEG studies

Differences have been identified between animal species for the sus-ceptibility to drug-induced seizure (Bassett et al., 2014). The rank orderbetween species for seizure susceptibility can differ between com-pounds but the Beagle dog is often the most susceptible (Elander,2013) possibly owing to genetic predisposition to idiopathic epilepsy(Edmonds et al., 1979). Cases for which the Beagle dog was less suscep-tible than other species were also reported (Authier, unpublished data)and careful and conservative interpretation of the preclinical data is al-ways warranted. Within an animal model, inter-individual differencescan be observed for anticonvulsant (Bankstahl, Bankstahl, & Löscher,2012; Brandt, Volk, & Löscher, 2004; Klein, Bankstahl, & Löscher,2015) and proconvulsant effects (Bassett et al., 2014; Himmel, 2008).The route of administration can also account for part of the variabilityobserved in EEG studies with oral dosing typically associated withgreater variations in exposures than parenteral dosing and consequent-ly increased CNS effect variability.

Decisions on the safety of drugs may be based on a broad range ofconsiderations including in silicomodeling, receptor affinity, drug expo-sure, histopathology and/or seizure characteristics (e.g. precursor EEGchanges, precursor clinical signs, seizure onset triggers, duration/self-limiting, success of emergency treatments in animals, recovery, kindlingeffects, inter-individual variability and incidence, species differences,etc.(Markgraf et al., 2014). In particular, clinical signs in animals maybe correlated with the exhibition of seizures and/or abnormal brain ac-tivity following drug administration (see Section 8 for further discus-sion). However, skepticism exists that the exhibition of these signs inanimals does not correlate with signs that are predictive of seizureand/or brain activity in humans. Therefore, a caveat remains that the se-quence and drug exposure concentrations at which these precursorsigns of seizure are observed in animal studies may differ from thehuman response. Precursor clinical signs of CNS toxicity are often non-specific (e.g. tremors, nystagmus, emesis, ataxia) and the mechanismsinvolved may also differ between species. While differences can be ob-served, animal models remain a cornerstone of drug safety testingprior to first in human administration. Similar precursor sign profilesare observed across species with pentylenetetrazol (PTZ), a commonlyused positive control (Bassett et al., 2014) but also for a broader rangeof pharmaceutical agents (Easter et al., 2009). Seizure liability studiesoften aim to determine the EEG NOEL (No Observed Effect Level) orNOAEL (NoObserved Adverse Effect Level) in the test system to later es-timate safe doses for clinical investigations. There is agreement among

the authors that plasma concentrations at doses that produce seizuresand/or abnormal brain activity in animals are likely correlated to thosethatmay produce the same effect in humans. Therefore, plasma concen-trations at the dose estimated as a NOAEL in animals may be used to es-tablish safetymargins, which are useful when developing a clinical doseescalation plan and PK dose-stopping criteria (see Section 8 for furtherdiscussion).

6. Seizure risk assessment: Data to consider when assessing seizurerisks

Seizure risk assessmentmay be based on various findings such as PKdata on the parent drug (and biologically active metabolites), clinicaland anatomical pathology findings, physiological alterations, and be-havioral observations. The PK profile(s) should include values for timeto peak concentration (i.e., Tmax), peak concentration (Cmax), overallsystematic exposure (AUC), and elimination half-life (t1/2). Values forTmax and t1/2 should be considered when evaluating the time coursefor EEG findings and premonitory signs of convulsions. This consider-ation may be helpful in determining which chemical is responsible forinducing seizures and other related toxicities based on the plasma levelsof each chemical at the time these effects occur. Seizures are known tobe induced following alterations in clinical pathology endpoints that in-clude sodium, calcium, andmagnesium. Also, anatomical changes in thebrain may induce convulsions and seizures (e.g., head injury). With re-gard to behavior, premonitory signs of convulsions in animals includehead and body tremors, incoordination, and ataxia (Bassett et al.,2014; Smythe, Ryan, & Pappas, 1988). Although these signs may be re-lated to convulsions, there are instances in which each may be due toother pharmacological effects produced by the product. For example,behaviors such as tremors and shaking may be due to hypothermia.Note that there is evidence that hyperthermia and hypothermiacan induce seizures in animals (Smythe et al., 1988; van Gassenet al., 2008; Kallman 2008). EEG can provide a definitive confirma-tion of the presence of seizure based on the observation of ictal activ-ity (i.e. EEG spike train). Clinically, there is no minimum time todefine a seizure and EEG discharges accompanied by clinical signsof seizure qualify as an electrographic seizure irrespective of theirduration (Fisher et al., 2014). Overall, the seizure risk assessmentshould consist of various types of data to determine if a seizure oc-curred and, if so, the exposure level of the chemical that inducedthis effect.

7. Safety margins: Factors to consider when estimating margins

Although there is an official FDA policy on how to determine safetymargins for the starting dose of initial clinical trials in adult healthy vol-unteers, the guidance that provides this information does not provideinsight on how to determine safety margins for dose-escalation treat-ment schedules (see the FDA guidance for industry: Estimating theMaximum Safe Starting Dose in Initial Clinical Trials for Therapeuticsin Adult Healthy Volunteers, available at http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm078932.pdf. The safety evaluation for each drug musttake into consideration the risks and benefits to the intended patientpopulation. In general, a safety margin of ≥10-fold is usually acceptablefor products that produce irreversible and unmonitorable adverse ef-fects (e.g. unexplained death, severe anatomical pathology findingssuch as liver necrosis, seizures). A few key factors to consider when es-tablishing safety margins include patient population, route of adminis-tration, and drug interactions. Patient populations with increasedvulnerability to convulsions include those with a history of seizuresand/or head injury (Kim et al., 2006; Schierhout and Roberts, 2012).Also, patient populations with a history of chronic use of alcohol andother drugsmaydecrease the seizure threshold and increase vulnerabil-ity to convulsions (Enevoldson, 2004;Hillbom, Pieninkeroinen, & Leone,

Page 8: Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive electrocardiogram (ECG)monitoringiscommonly included inpreclin-icalsafety studies,pre-doseorpost-doseEEGassessments

281S. Authier et al. / Journal of Pharmacological and Toxicological Methods 81 (2016) 274–285

2003). The age of the studypopulation should also be taken into accountsince seizure vulnerability may be increased in younger patients due tothe underdevelopment of detoxification systems and in older patientsgiven the inefficiency of the same systems. In rodents, findings demon-strate that spontaneous seizures (e.g., spike wave seizures) increase infrequency as animals age, which provides further evidence that theage of subjects is a critical variable in evaluating seizures (Ellens et al.,2009; Kelly, 2010; Sgro, Benagh, Modlin, & Kallman, 2006; Sitnikova,Hramov, Grubov, & Koronovsky, 2014). The route of administration isconsidered since the absorption of the product may vary dependingon the route employed. For example, drugs administered intravenously(IV) are entirely bioavailable (i.e. 100%), whereas those that are admin-istered via other routes may not be. The absorption of drugs via theseother routes may be highly variable across patients, which adds uncer-tainty to themargin needed to ensure safety. In regard to potential druginteractions, safetymargins greater than 10-foldmay need to be appro-priatewhen the test product is co-administeredwith a proconvulsant orconvulsant agent.

8. Examples when nonclinical EEG studies can help determine safeclinical exposure levels

Drugs effects may represent biological responses that occur sponta-neously in organisms and that overlap temporally. Also, the potency inwhich a drug (and its metabolites) produces an effect may vary acrossspecies, resulting in the observation of different levels of sensitivity tothe drug. These issues complicate the interpretation of findings acrossanimal species in which convulsions and abnormal brain activity (in-cludes seizures) are observed. Therefore, the scenarios discussedbelow illustrate the use of EEG techniques to determinewhether clinicalsigns and convulsions were due to abnormal brain activity that is drug-related and to establish the most sensitive and/or relevant species. Thegoal of any toxicology study is to define doses that are appropriate tostudy in humans and to avoid unwanted side effects. This is completedby both defining a NOEL/NOAEL and the exposure levels at which unac-ceptable adverse effects occur. The scenarios below illustrate the use ofexposure levels in the most sensitive and/or relevant species to estab-lish PK dose-stopping criteria for clinical studies. In the scenariosdiscussed, EEG techniques are used to establish exposure levels atwhich abnormal brain activity (includes seizures) is not observed at ei-ther the NOEL or NOAEL estimated in animals. These levels are used toestablish PK dose-stopping criteria that set the top clinical exposurelevels (i.e., for Cmax and/or AUC) at 1/10 that measured at either theNOEL or NOAEL estimated in the most sensitive and/or relevant animalspecies. This approach is used given the nature of the toxicity of concern(abnormal brain activity) and to help ensure that the toxicity is not ob-served in patients, especiallywhen the benefits of treatment do not out-weigh the risks associated with it. Note that the scenarios do notrepresent findings from an actual study report. These scenarios werewritten to illustrate challenges thatmay arisewhen evaluatingpotentialtherapeutics that may be seizurogenic agents that do not significantlyalter clinical pathology endpoints. The conclusions provided are fromthe perspective of the authors and represent one of many approachesto interpreting the findings provided.

As mentioned above, convulsions may occur spontaneously in ani-mals. EEG studies offer a definitive means to determine if clinical signssuch as convulsions or other premonitory signs are due to abnormalbrain activity or not. This can be complicated by the background inci-dence of convulsions in the species tested. For example, Drug A (0, 20,100, or 500 mg/kg) is administered IV to dogs in a single-dose toxicolo-gy study. DrugA is known to produce hypothermia and sedation inmul-tiple species. In the current study, dogs exhibit hypoactivity at≥100 mg/kg; tremors across all doses; and a convulsion at 500 mg/kg(high dose). The convulsion is exhibited in a dog that exhibited tremors.Given the observation of tremors in this animal it is considered a poten-tial premonitory sign of convulsions in the low and mid dose groups.

Therefore, a NOAEL cannot be established since a convulsion was ob-served at the high dose and tremors are exhibited across the doses test-ed. This scenario is challenging since the incidence of convulsions waslow and in a species known to exhibit spontaneous convulsions(Bielfelt et al., 1971) and the tremors observed may have simply beensecondary to hypothermia. This information suggests that the convul-sionmay not be drug related andwarrants further EEG testing. In a sep-arate study, drug naive dogs exhibit tremors, in the absence of abnormalEEG activity at the doses previously tested. The convulsion is notreproduced in a second study and EEG data suggest no evidence of ab-normal brain activity in the presence of premonitory signs. These datasupport the conclusion that the convulsion observed in the initialstudy may have been spontaneous given its low incidence and thelack of abnormal EEG activity in treated dogs.

In scenarios in which abnormal brain activity is detected via EEGtechniques, PK dose- stopping criteria can be established for the clinicbased on plasma levels of drug in the most sensitive species. For exam-ple, Drug B is orally administered to rats (0, 1, 10, or 17mg/kg) and dogs(0, 0.5, 2.5, or 5mg/kg). Based on PK data, the oral bioavailability of DrugB ranges from 10 to 70% and the excretion of unchanged drug in urinesamples appears complete (100%) across species. Both species exhibitconvulsions at the highest dose tested. Given the observation of shakingand uncoordinated behavior at the high dose group both signs wereconsidered premonitory signs of convulsion in animals from the low-and mid-dose groups that exhibit them. A NOAEL cannot be estimatedin either species since convulsions are observed at the high dose andpremonitory signs are exhibited across the doses tested. The highly var-iable oral bioavailability confounds the determination of themost sensi-tive species based on body surface area comparisons. As a follow-up,Drug B (same doses) is evaluated in rats and dogs using EEG techniquesin order to correlate the findings with exposure data. In rats, abnormalEEG activity is observed at the high dose and characterized by the ap-pearance of high frequency gamma activity, repetitive bursts of sharpwaves, and increased sharp waves. In dogs, abnormal EEG activity is ob-served at the mid- and high-dose and is characterized by the appear-ance of high frequency gamma activity at the high dose and repetitivebursts of sharp waves and increased sharp waves at the mid and highdose. Across species, abnormal EEG activity is observed at 1 h followingtreatment, which correlates with the averaged Tmax value for Drug B.There is no evidence of frank seizure in either species at the doses test-ed. A NOEL can be estimated in rats and dogs, respectively, at 10 mg/kgand 0.5mg/kg based on the lack of abnormal EEG activity at these doses.The use of EEG techniques provides a definitive evaluation of alterationsin CNS activity, which in turn allows for a more informed decision as towhether or not the premonitory signs exhibited are associated with ab-normal brain activity. Overall, the dog is deemed themost sensitive spe-cies given that the Cmax value at its NOEL (333 ng/mL) was lower thanthat in rats (555 ng/mL).

PK dose-stopping criteria can be established based on the NOEL indogs, given the nature of the toxicities that define it. The PK dose-stopping criteria establish the top Cmax value allowed in the clinic. Thetop value (33.3 ng/mL) is set 10-fold lower than the exposure level atthe NOEL in the dog given the marked variability in the oral absorptionof the drug and the nature of the toxicity of concern (i.e., abnormal brainactivity). This scenario illustrates that premonitory signs such as shak-ing and uncoordinated activitymay be indicative of abnormal EEG activ-ity in animals; however, these signs do not always correlate with EEGabnormalities. Also, findings in the dog demonstrate that abnormalEEG activity occurs at exposure levels of drug that are lower thanthose at which frank convulsions occur.

EEG data can also be useful in evaluating drugs with a different met-abolic profile in the test species compared to humans, a scenario that isnot always straightforward in terms of how to appropriately establishsafety margins to inform clinical dose-escalation limitations. As such,being able to correlate the appearance of abnormal EEG activity withthe Tmax of a parent or major metabolite can provide valuable data to

Page 9: Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive electrocardiogram (ECG)monitoringiscommonly included inpreclin-icalsafety studies,pre-doseorpost-doseEEGassessments

Fig. 9. Cmax plasma levels for preclinical toxicology data — a compound that producestremor, seizures and brain lesions.

282 S. Authier et al. / Journal of Pharmacological and Toxicological Methods 81 (2016) 274–285

assist in the design of the proposed clinical study. For example, Drug C isorally administered to monkeys, dogs, and rats. Convulsions are ob-served in these species following the administration of Drug C, whichis known to form a biologically active metabolite M123. Comparisonof the drug/metabolite ratio in the nonclinical test species to humansmay help determine which animal species may be the most relevantIn this example, the metabolite was formed in dogs and monkeys, butnot rats and themetabolic profile in themonkeymost closely resemblesthat of humans. As all species exhibited convulsions, theparent is clearlycontributing to the convulsions, however, the role of the metabolite, ifany, in the convulsions noted is still not clear.

As a follow-up, Drug C and the isolatedM123metabolite is evaluatedinmonkeys using EEG techniques to determine if abnormal EEG activityis induced by one or both compounds. Evidence of frank seizure and ab-normal EEG activity is observed inmonkeys at themid and high dose ofDrug C, confirming that the observed convulsions are indeed seizures.Frank seizure and abnormal EEG activity are also observed followingtreatment with the M123 alone, demonstrating that both compoundsmust be considered seizurogenic. A NOEL for Drug C is estimated at2mg/kg (Cmax= 22 ng/mL) based on evidence of abnormal EEG activ-ity and frank seizure at higher doses. A NOEL could not be establishedfor M123 given the abnormal EEG activity and frank seizure observedat the dose tested. However, these data provide reasonable justificationthat the monkey can be used to establish real-time PK dose-stoppingcriteria based on the parent exposure levels alone, given the compara-bility of the metabolic profile with humans. Therefore, the top clinicalCmax was established at 2.2 ng/mL for the parent drug as this plasmalevel takes into account both the parent and metabolite levels. If themetabolic profile was not comparable to humans, stopping criteriawould have to take into consideration both the parent and themetabo-lite Cmax.

This scenario illustrated that the most relevant animal species maybe determined based on the metabolite profile in the species tested,the isolated metabolite should be tested alone to evaluate its effectson EEG activity, and the PK stopping criteria should include top expo-sure levels for the parent drug and metabolites demonstrated to pro-duce abnormal EEG activity and other adverse effects. A riskassessment should be determined for both the parent drug and its bio-logically active metabolites. This assessment should be based on find-ings from studies that evaluate the parent drug and its biologicallyactive metabolites alone and in combination. Doses of the metaboliteshould be selected to ensure exposure levels comparable to that ob-served following treatment with the doses of parent drug tested inorder to ensure the evaluation of relevant exposure levels. The Tmax

values for both the parent and its metabolites should be compared tothe time points in which adverse effects such as abnormal EEG activityare observed in order to understand the potential role of these chemicalentities in the effects measured following treatment. The interaction ofthe parent drug and its metabolite may be complex and result in differ-ent pharmacological and toxicological profiles. Therefore, these resultsmust be carefully interpreted, especially when used to establish PKdose-stopping criteria.

9. Translation of non-clinical EEG to the clinic

Translation of EEG findings to the clinical environment makes thecritical impact on the regulatory environment. Other measures of con-vulsive alterations such as proconvulsive assessments, where thenovel new drug is combined with a known convulsant like picrotoxin,kainic acid, electroshock, pentylenetetrazol, or prior electrical kindlingto determine changes in threshold are useful preclinical tools that lackdirect translation to the clinic. An indirect ranking of the convulsive lia-bility of compounds may be possible but these same paradigms are notconducted with human subjects since all of them depend on establish-ing convulsion prior to adding the new drug to the paradigm. Essential-ly, how can these preclinical assessments support human safety and the

design of clinical evaluations of newpharmaceuticals? The primary pre-clinical assessment tool that can bemonitored in humans is EEG. Severalfactors must be considered in the quality of preclinical EEG for humantranslation and safety evaluations. The major issues that must be con-sidered are species differences, age differences in EEG, EEG assessmenttools, and the limitations of predictability from preclinical assessmentto the clinical environment.

The first approach in understanding translation is making a distinc-tion between reported observations of convulsion from seizures or thealteration in the electrical activity patterns distinguished from EEG re-cordings. Typically the goal would be to avoid convulsion by establish-ment of a same dose or identification of premonitory effects thatwould aid in the avoidance of convulsive doses of the new compound.For a premonitory event to be useful in managing a clinical drug trialthe event must occur at lower doses that do not produce convulsions/seizures or be separated from the convulsive/seizure event by a long pe-riod of time. Potential premonitory events cannot be an aura to the sei-zure which indicates that the seizure is eminent since these aura eventsdo not provide sufficient time to abate the seizure. Fig. 9 illustrates howclinical signs, other toxicity indicators, and plasma drug level can beused to put a seizure effect into perspective. Timeframe can be an im-portant variable for monitoring delayed onset seizures with repeateddosing and the data generation approach tomonitor delayed onset con-vulsions and Cmax driven convulsions would be quite different. SeeTable 2 for a summary of differences between acute and chronic EEGmonitoring methods. For the delayed onset events, EEG in the non-precipitated situationmay change overtime prior to the onset of seizureevents (Carfagna, Sgro, Arezzo, & Kallman, 2010; Kallman, 2006). Alter-ations in muscular endpoints including myoclonus (Kojovic, Cordivari,& Bhatia, 2011) can be easily defined as convulsion by observers. Incor-rect labeling of thesemuscular events as convulsion is frequent and canonly be corroborated by EEGmonitoring. The distinction betweenmus-cular changes and actual EEG spike activity dramatically affect the clin-ical understanding of these effects. Similarly, drug induced catalepsymay be incorrectly defined as convulsion (Bricker, Sampson, &Ablordeppey, 2014; Heitz & Bence, 2013; Sanberg, Bunsey, Giordano,& Norman, 1988; Winters, Ferrar-Allado, Guzman-Flores, & Alcaraz,1972), again with EEG serving as a key biomarker for proper identifica-tion of the drug induced effect.

Once a true seizure profile has been identified the finding should beput into perspective to improve clinical safety. Many drugs used thera-peutically have seizure profiles at therapeutic doses (Alper, Schwartz,Kolts, & Khan, 2007). Typically the seizure profile for clinical compoundsis observed at the higher doses, with long term use of the drug, and inpatients with a history or predisposition to seizure prior to drug admin-istration. The most critical factors to understand for translation is themargin of the seizurogenic effect to the therapeutic dose, establishinga NOEL/NOAEL, understanding the pre-seizure events that would be re-liably monitored, the therapeutic endpoint, and whether the effect isCmax driven or has a delayed onset. All of these preclinical variableswould impact the design of clinical testing and the probability of drugdevelopment success in the clinic.

The consistency of the observation of convulsions/seizures should beevaluated across species. Sometimes a compound has a unique

Page 10: Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive electrocardiogram (ECG)monitoringiscommonly included inpreclin-icalsafety studies,pre-doseorpost-doseEEGassessments

Table 2Comparison of preclinical acute and chronic EEG recordings.

Acute EEG Chronic EEG

▶ No surgical implantation ▶ Surgical implantation/surface or deepelectrodes

▶ No mapping of sites ▶ Can map sites▶ Restrained animal ▶ No restraint▶ Short duration recording ▶ 24/7 up to 3 months or more▶ Identification of seizure activity/-

spiking no qEEG or staging▶ Identification of seizure activity/spiking,

pharmaco-EEG, qEEG, staging▶ More diagnostic rather than time

expansive▶ Focus on time expansive recordings

Table 3Pros and cons of using screening EEGs on large animal studies.

Advantages of EEG Prescreens Disadvantages of EEG Prescreens

• Dogs may have a high rate of convul-sion at the time of receipt fromsuppliers – Reported as 4% (Arezzo2005) and as 0.02% based on the ven-dor database

• Not as useful in rodents where the inci-dence is lower (less than 1%) and rodentstrain is an issue

• Can be incorporated into an ongoingtoxicology study

• May have to conduct additional studiesto understand special populationsensitivity

• Few Ns in large animal toxicologystudies (typically 3–6/treatmentgroup)

• Although humans are typically notprescreened they could be to establish aclinical safety margin

• Special population preclinical datacan be obtained later in drugdevelopment, if required, in a sepa-rate study

• May have to understand later issues relat-ed to special sensitivity of therapeuticpopulation since humans on later clinicaltrials may be less homogeneous than ani-mal population

• Early goal is to understand thresholdor NOAEL under consistent conditions

• Additional cost to toxicology studies –may require additional animals forexclusion

• Consistency of population for makingdecisions for early healthy subjectsafety trials

• Must develop exclusion criteria

• Prescreens for other endpoints ontoxicology studies is based on the ideaof healthy animals concept

• Not a good prescreen requirement forall toxicology studies but rather forcause or expected profile

• Can distinguish changes due to drugexposure in non-epileptic animals

• Cannot detect more subtle qEEG chang-es but seizure activity only inprescreens

• Initial Phase 1 clinical studies are fo-cused on normal or healthy clinicalpopulation

• Might be incorrectly applied when in-terested in EEG effects other thanthreshold determinations

• Supportive of studies designed to de-termine if observational convulsionsare seizures or due to motoric chang-es. Baseline EEG must be normal tomake this determination

• Requires high quality expertise

Table 4Differences between preclinical and clinical EEG recordings.

Preclinical Clinical

• Acute recordings in restrainedanimals

• Holter monitoring and acute scalp elec-trodes in resting state

• Telemetry recordings in surgicallyprepared animals

• Multiple sites

• Usually 1–2 general sites on brainsurface

• Limited to therapeutic doses for evaluation

• Deep electrode for evaluation ofhippocampal theta in sleepevaluation

• Video-EEG considered as gold standard

• Can evaluate dose higher thantherapeutic doses

• Coincidental EMG for signs not possible

• Can record simultaneous video andEMG

• Confidence in identification of seizure/pre--seizure activity but less confidence in qEEG.

283S. Authier et al. / Journal of Pharmacological and Toxicological Methods 81 (2016) 274–285

metabolite profile (Sullivan, Hanasono, Miller, & Wood, 1987) thatcould be associated with convulsive behavior. Most toxicology pro-grams include a rodent and large animal species which is usually thedog. As one ascends the phylogenetic scale there aremore sophisticatedmechanisms to inhibit seizures as a result of increased cortical inhibi-tion and enhanced reduction in the spread of electrical excitation fromhemisphere to hemisphere characteristic of convulsive profiles. An ob-servation of convulsion in the rat may not be corroborated in the dogor primate (Kallman, Sgro, Markgraf, & Ballering, 2014). The primatebrain, organization of the cortex, corpus callosum and descending regu-lation of CNS stimulation is most like the human (Katz, Lasek, & Silver,1983) but many times not the species of choice for more detailed toxi-cology evaluations where pharmacokinetics, metabolite profile, andother toxicological effects are well described in GLP studies. As previ-ously discussed, the dog offers some special considerations since thisspecies is frequently selected for characterizing toxicity but is recog-nized to present a higher rate of seizure profiles prior to drug treatment(Bielfelt et al., 1971). Some laboratories have resorted to prescreeningdogs prior to the start of toxicology studies where there is an indicationthat convulsion/seizure is a potential safety issue. These are conductedon restrained dogs by a short (e.g. 2–10 min) acute EEG recordingwith surface needle electrodes to determine if seizure activity or abnor-mal EEG morphologies is present. Table 3 lists the major pros and consof prestudy screening applied to non-clinical studies. The applicationto the toxicology study, i.e. pre-FHD or pre-Phase 3 clinical testing,may impact the decision to apply prescreening.

A final consideration is to contrast data from preclinical to clinicalEEG evaluations. Table 4 lists the technical differences between the an-imal and human approaches for recording. The two approaches can pro-vide different levels of sensitivity for detection with the humanapproach generally considered as more sophisticated and often of lon-ger duration since a continuous recording approach is commonlyused. When the focus is on seizure activity and not qEEG characteriza-tion the human and animal data are more consistent and less variablethan when qEEG characterization is of interest.

10. Discussion

EEG appears to be underutilized in non-clinical drug safety testing.Epidemiological considerations weight for an increasing importance ofCNS safety issues owing to the aging population and growing preva-lence of polypharmacy. Seizure and sleep liabilities are manifest appli-cations of EEG to safety testing of drug candidates but challengesremain in the use of this methodology including data interpretation,translation from animals to the clinic and drug safety profile variants.Interpretation challenges originate from the abundance of data butalso from the inherent diversity of EEGmorphologies that can be obtain-ed.Mechanisms to explain drug induced adverse effects are often poorlycharacterized which adds to the uncertainty during seizure risk assess-ment using animal data. As discussed, thorough characterization ofpharmacokinetics can inform species comparisons with the goal ofpredicting thehuman response. Someagents induce adverse CNS effectsafter acute administration while others require chronic exposure.

Tachyphylaxis, a decrease in the response to a drug, is commonly ob-servedwith CNS active drugs andmay impact the design of EEG studies.During toxicology studies, tremors (involuntary symmetrical oscilla-tions of a body part), myoclonus (brief, involuntary muscle jerk causedby abrupt muscle contraction) and convulsions (abnormal, violent andinvoluntary contraction or series or contractions of the muscles) maybe observed. Each of these observations may be associated with EEGbiomarkers of increased seizure risk or frank ictal activity. EEGmonitor-ing is essential for proper characterization of the clinical events that areobserved. As a well-established diagnostic methodology, EEG monitor-ing has the potential to offer a high translational value but a complexmatrix of scientific and regulatory considerations is involved duringdecision-making leading to the inclusion of this neurological monitor-ing endpoint in preclinical studies. From design to interpretation, the

Page 11: Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive electrocardiogram (ECG)monitoringiscommonly included inpreclin-icalsafety studies,pre-doseorpost-doseEEGassessments

284 S. Authier et al. / Journal of Pharmacological and Toxicological Methods 81 (2016) 274–285

drug development team is faced with multiple aspects to integrate asthey exercise the art of safety pharmacology.

Conflict of interest

None of the authors have any conflicts of interest, other than theiremployment in commercial pharmaceutical companies, academic insti-tutions or contract research organizations. No information is presentedin this paper that advocates for or promotes commercial products fromany of our organizations.

Disclaimer

This publication reflects the views of the authors and does not repre-sent views or policies of the any organization, including the FDA. Theviews of the authors should not be construed to represent FDA's viewsor policies.

Acknowledgment

The authorswish to thank the Safety Pharmacology Society (SPS) forsupport in the conduct of the Continuing Education course conducted atthe 2014 Annual Safety PharmacologyMeeting thatwas the basis of thismanuscript.

References

Acharya, U. R., Sree, S. V., Swapna, G., Martis, R. J., & Suri, J. S. (2013). Automated EEG anal-ysis of epilepsy: A review. Knowledge-Based Systems, 45, 147–166.

Alper, K., Schwartz, K. A., Kolts, R. L., & Khan, A. (2007). Seizure incidence in psychophar-macological clinical trials: an analysis of food and drug administration (FDA) summa-ry basis of approval reports. Biological Psychiatry, 62, 345–354.

Authier, S., Arezzo, J., Delatte, D. S., Kallman, M. J., Markgraf, C., Paquette, D., ... Curtis, M. J.(2016). Safety pharmacology investigations on the nervous system: An industry sur-vey. Journal of Pharmacological and Toxicological Methods, 81, 37–46.

Authier, S., Bassett, L., Pouliot, M., Rachalski, A., Troncy, E., Paquette, D., & Mongrain, V.(2014). Effects of amphetamine, diazepam and caffeine on polysomnography (EEG,EMG, EOG)-derived variables measured using telemetry in cynomolgus monkeys.Journal of Pharmacological and Toxicological Methods, 70(1), 86–93.

Authier, S., Paquette, D., Gauvin, D., Sammut, V., Fournier, S., Chaurand, F., & Troncy, E.(2009). Video-electroencephalography in conscious non-chuman primate using ra-diotelemetry and computerized analysis: Refinement of a safety pharmacologymodel. Journal of Pharmacological and Toxicological Methods, 60(1), 88–93.

Authier, S., Schaeffer, E., Weed, M., Digavalli, S., Maghezzi, S., Gervais, J., ... Fournier, S.(2012). Arousal and spectral analysis: EEG in conscious cynomolgus monkeys.Journal of Pharmacological and Toxicological Methods, 66, 164.

Azzam R, Bhatt AB. (2014). Mimickers of generalized spike and wave discharges. TheNeurodiagnostic Journal .54(2):156–62.

Bankstahl, M., Bankstahl, J. P., & Löscher, W. (2012). Inter-individual variation in the an-ticonvulsant effect of phenobarbital in the pilocarpine rat model of temporal lobe ep-ilepsy. Experimental Neurology, 234(1), 70–84.

Bassett, L., Troncy, E., Pouliot, M., Paquette, D., Ascah, A., & Authier, S. (2014). Telemetryvideo-electroencephalography (EEG) in rats, dogs and non-human primates:Methods in follow-up safety pharmacology seizure liability assessments. Journal ofPharmacological and Toxicological Methods, 70(3), 230–240.

Benbadis, S. R., & Tatum, W. O. (2003). Overintepretation of EEGs and misdiagnosis of ep-ilepsy. Journal of Clinical Neurophysiology, 20(1), 42–44.

Bielfelt, S.W., Redman, H. C., &McClellan, R. O. (1971). Sire- and sex-related differences inrates of epileptiform seizures in a purebred beagle dog colony. American Journal ofVeterinary Research, 32(12), 2039–2048.

Brandt, C., Volk, H. A., & Löscher, W. (2004). Striking differences in individual anticonvul-sant response to phenobarbital in rats with spontaneous seizures after status epilep-ticus. Epilepsia, 45(12), 1488–1497.

Bricker, B., Sampson, D., & Ablordeppey, S. Y. (2014). Evaluation of the potential of anti-psychotic agents to induce catalepsy in rats: Assessment of a new, commerciallyavailable, semi-automated instrument. Pharmacology, Biochemistry, and Behavior,120, 109–116.

Bruse, D. K., Zinter, E., Brooks, K., Valderas, M., Wilder, J., Hutt, J., ... Lyons, R. (2012). Uti-lization of telemetry to characterize physiological changes following F. tularensisSCHU S4 aerosol in unvaccinated and vaccinated cynomolgus macaques. Journal ofPharmacological and Toxicological Methods, 66(2), 194–195.

Carfagna, M., Sgro, M. P., Arezzo, J. C., & Kallman, M. J. (2010). Translation of seizure poten-tial during treatment with anmGlu2/3 agonist prodrug (LY2140023). Society of Toxicol-ogy Presentation.

Edmonds, H. L., Jr., Hegreberg, G. A., van Gelder, N. M., Sylvester, D. M., Clemmons, R. M., &Chatburn, C. G. (1979). Spontaneous convulsions in beagle dogs. FederationProceedings, 38, 2424–2428.

Elander, M. (2013). Drug-induced convulsions in nonclinical safety studies: Implicationfor clinical development. Drug Development Research, 74(3), 155–161.

Ellens, D. J., Hong, E., Giblin, K., Singleton, M. J., Bashyal, C., Englot, D. J., ... Blumenfeld, H.(2009). Development of spike-wave seizures in C3H/HeJ mice 1. Epilepsy Research, 85,53–59.

Enevoldson, T. P. (2004). Recreational drugs and their neurological consequences. Journalof Neurology, Neurosurgery, and Psychiatry, 75(Suppl. 3), iii9–ii15.

Engel, J., Bragin, A., Staba, R., & Mody, I. (2009). High frequency oscillations: What is nor-mal and what is not? Epilepsia, 50, 598–604.

Easter, A., Bell, M. E., Damewood, J. R., Jr, Redfern, W. S., Valentin, J. P., Winter, M. J., ...Bialecki, R. A. (2009). Approaches to seizure risk assessment in preclinical drug dis-covery. Drug Discov Today, 14(17–18), 876–884.

Ferguson, J. M. (2001). SSRI antidepressant medications: Adverse effects and tolerability.The Primary Care Companion to The Journal of Clinical Psychiatry, 3(1), 22–27.

Fisher, R. S., Scharfman, H. E., & deCurtis, M. (2014). How can we identify ictal andinterictal abnormal activity? Advances in Experimental Medicine and Biology, 813,3–23.

Fisher, Scharfman, & Decurtis (2014). Advances in Experimental Medicine and Biology, 813,3–23.

Fox, S. V., Gotter, A. L., Tye, S. J., Garson, S. L., Savitz, A. T., Uslaner, J. M., ... Renger, J. J.(2013). Quantitative electroencephalography within sleep/wake states differentiatesGABAA modulators eszopiclone and zolpidem from dual orexin receptor antagonistsin rats. Neuropsychopharmacology, 38, 2401–2408.

Galanopoulou, A. S., Kokaia, M., Loeb, J. A., Nehlig, A., Pitkänen, A., Rogawski, M. A., et al.(2013). Epilepsy therapy development: Technical and methodologic issues in studieswith animal models. Epilepsia, 54(Suppl. 4), 13–23.

Heitz, C. R., & Bence, J. R. (2013). Ketamine-induced catalepsy during adult sedation in theemergency department. The Journal of Emergency Medicine, 44(2), e243–e245.

Hillbom, M., Pieninkeroinen, I., & Leone, M. (2003). Seizures in alcohol-dependent pa-tients: epidemiology, pathophysiology and management. CNS Drugs, 17(14),1013–1030.

Himmel, H. M. (2008). Safety pharmacology assessment of central nervous system func-tion in juvenile and adult rats: Effects of pharmacological reference compounds.Journal of Pharmacological and Toxicological Methods, 58(2), 129–146.

Iber, C., Ancoli-Israel, S., Chesson, A. L., & Quan, S. F. (2007). The AASM manual for the scor-ing of sleep and associated events. American Academy of Sleep Medicine. IL:Westchester.

Irvine, R. J., White, J., & Chan, R. (1997). The influence of restraint on blood pressure in therat. Journal of Pharmacological and Toxicological Methods, 38(3), 157–162.

Ivarsson, M. (2009). Sleep changes as translational pharmacodynamic biomarkers.European Pharmaceutical Review, 5, 40–44.

Jasper, Herbert H. (1958). Report of the committee on methods of clinical examination inelectroencephalography: 1957. Electroencephalography and Clinical Neurophysiology,10(2), 370–375.

Jobert, M., Wilson, F. J., Ruigt, G. F., Brunovsky, M., Prichep, L. S., & Drinkenburg, W. M.(2012). Guidelines for the recording and evaluation of pharmaco-EEG data inman. The International Pharmaco-EEG Society (IPEG). Neuropsychobiology,66(4), 201–220.

Kallman, M. (2006). Reducing adverse clinical risk for convulsion by understanding premon-itory events and potential biomarkers. Society of Toxicology Presentation.

Kallman, M., Sgro, M., Markgraf, C., & Ballering, M. (2014). When a convulsion is not a sei-zure: behavioral and EEG studies with SCH A in the beagle dog. Poster presented at So-ciety of Toxicology.

Katz, M. J., Lasek, R. J., & Silver, J. (1983). Ontophyletics of the nervous system: Develop-ment of the corpus callosum and evolution of axon tracts. Proceedings of theNational Academy of Sciences, 80, 5936–5940.

Kelly, K. M. (2010). Agingmodels of acute seizures and epilepsy. Epilepsy Currents, 10, 15–20.Kim, L. G., Johnson, T. L., Marson, A. G., Chadwick, D.W., &MRCMESS Study group (2006).

Prediction of risk of seizure recurrence after a single seizure and early epilepsy: Fur-ther results from the MESS trial. Lancet Neurology, 5(4), 317–322.

Klein, S., Bankstahl, M., & Löscher, W. (2015). Inter-individual variation in the effect of an-tiepileptic drugs in the intrahippocampal kainate model of mesial temporal lobe ep-ilepsy in mice. Neuropharmacology, 90, 53–62.

Kojovic, M., Cordivari, C., & Bhatia, K. (2011). Myoclonic disorder: A practical approach fordiagnosis and treatment. Therapeutic Advances in Neurological Disorders, 4(1), 47–62.

Krauss, G. L., Abdallah, A., Lesser, R., Thompson, R. E., & Niedermeyer, E. (2005). Clinicaland EEG features of patients with EEG wicket rhythms misdiagnosed with epilepsy.Neurology, 14;64(11), 1879–1883.

Leiser, S. C., Dunlop, J., Bowlby, M. R., & Devilbiss, D. M. (2011). Aligning strategies forusing EEG as a surrogate biomarker: A review of preclinical and clinical research.Biochemical Pharmacology, 81, 1408–1421.

Markgraf, C. G., DeBoer, E., Zhai, J., Cornelius, L., Zhou, Y. Y., & MacSweeney, C. (2014). As-sessment of seizure liability of Org 306039, a 5-HT2c agonist, using hippocampalbrain slice and rodent EEG telemetry. Journal of Pharmacological and ToxicologicalMethods, 70(3), 224–229.

Nagayama, T. (2015). Adverse drug reactions for medicine newly approved in Japan from1999 to 2013: Syncope/loss of consciousness and seizures/convulsions. RegulatoryToxicology and Pharmacology, 72(3), 572–577.

Nidermeyer, E., & Croft, J. R. (1961). Uber die bedeutung der 14 and 6 per sec positivenspitzen im EEG. Archiv für Psychiatrie und Nervenkrankheiten, 202, 266–280.

Nunn, G., & Macpherson, A. (1995). Spontaneous convulsions in Charles RiverWistar rats.Laboratory Animals, 29(1), 50–53.

Panayiotopoulos, C. P. (1999). Typical absence seizures and their treatment. Archives ofDisease in Childhood, 81(4), 351–355.

Paterson, L. M., Nutt, D. J., & Wilson, S. J. (2011). Sleep and its disorders in translationalmedicine. Journal of Psychopharmacology, 25, 1226–1234.

Page 12: Journal of Pharmacological and Toxicological Methods · changes insleep.Yet, whilenon-invasive electrocardiogram (ECG)monitoringiscommonly included inpreclin-icalsafety studies,pre-doseorpost-doseEEGassessments

285S. Authier et al. / Journal of Pharmacological and Toxicological Methods 81 (2016) 274–285

Pouliot, M., Bassett, L., Troncy, E., Abtout, S., Ascah, A., & Authier, S. (2015). Jacketed exter-nal electroencephalographic (EEG) telemetry monitoring in conscious beagle dogsand cynomolgus monkeys: Qualification of a central nervous system safety testingmodel. Journal of Pharmacological and Toxicological Methods, 75, 162–163.

Rachalski, A., Authier, S., Bassett, L., Pouliot, M., Tremblay, G., & Mongrain, V. (2014). Sleepelectroencephalographic characteristics of the cynomolgus monkey measured by te-lemetry. Journal of Sleep Research, 23(6), 619–627.

Rechtschaffen A, Kales A. (eds) (1977). Amanual of standardized terminology, techniquesand scoring system for sleep stages of human subjects. Los Angeles: UCLA, Brain In-formation/Brain Research Institute 1968

Reiher, J., & Lebel, M. (1977).Wicket spikes: clinical correlates of a previously undescribedEEG pattern. The Canadian Journal of Neurological Sciences, 4(1), 39–47.

Sanberg, P. R., Bunsey, M. D., Giordano, M., & Norman, A. B. (1988). The catalepsy test: Itsups and downs. Behavioral Neuroscience, 102(5), 748–759.

Satomoto, K., Kuroiwa, Y., Masubuchi, Y., Uemura, H., Oshima, Y., & Okazaki, S. (2012).Spontaneous convulsions in Sprague–Dawley rats in carcinogenicity studies. TheJournal of Toxicological Sciences, 37(3), 645–647.

Schierhout, G., & Roberts, I. (2012 Jun 13). WITHDRAWN: Antiepileptic drugs forpreventing seizures following acute traumatic brain injury. Cochrane Database ofSystematic Reviews, 6, CD000173.

Sgro, M., Benagh, V. J., Modlin, D. L., & Kallman, M. J. (2006). Age dependent alterations inpentylenetzol and electroshock convulsive thresholds in mice. Poster presented atNeurobehavioral Teratology Society Annual Meeting.

Sitnikova, E., Hramov, A. E., Grubov, V., & Koronovsky, A. A. (2014). Age-dependent in-crease of absence seizures and intrinsic frequency dynamics of sleep spindles inrats. The Journal of Neuroscience, 2014, 370764.

Smythe, J. W., Ryan, C. L., & Pappas, B. A. (1988). A behavioral and electrocorticographiccomparison of diazepam and pentylenetetrazol in rat pups. Pharmacology,Biochemistry, and Behavior, 30, 479–482.

Sterman MB. Power spectral analysis of EEG characteristics during sleep in epileptics.Pilepsia. 1981Feb;22(1):95–106.

Stoller, M. K. (1994 Sep-Octt). Economic effects of insomnia. Clinical Therapeutics, 16(5),873–897 (discussion 854).

Sullivan, H. R., Hanasono, G. K., Miller, W. M., & Wood, P. G. (1987). Species specificity inthe metabolism of nabilone. Relationship between toxicity and metabolic routes.Xenobiotica, 17(4), 459–468.

Toth, L. A., & Bhargava, P. (2013). Animalmodels of sleep disorders. Comparative Medicine,63(2), 91–104.

U.S. Food and Drug Administration (2001). Guidance for industry: Safety pharmacologystudies for human pharmaceuticals (S7A). http://www.fda.gov/cber/gdlns/ichs7a071201.htm

U.S. Food and Drug Administration (2010). Guidance for industry M3(R2)nonclinical safety studies for the conduct of human clinical trials andmarketing authorization for pharmaceuticals. http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm073246.pdf

van Gassen KL, Hessel EV, Ramakers GM, Notenboom RG, Wolterink-Donselaar IG,Brakkee JH, Godschalk TC, Qiao X, Spruijt BM, van NO and de Graan PN (2008). Char-acterization of febrile seizures and febrile seizure susceptibility in mouse inbredstrains. Genes, Brain, and Behavior, 7:578–586.

Veasey, S. C., Valladares, O., Fenil, P., Kapfhamer, D., Sanford, L., Benington, J., & Bucan, M.(2000). An automated system for recording and analysis of sleep in mice. Sleep, 23,1025–1040.

Vélez, L., & Selwa, L. M. (2003). Seizure disorders in the elderly. American Family Physician,67(2), 325–332.

Winrow, C. J., Gotter, A. L., Cox, C. D., Doran, S. M., Tannenbaum, P. L., Breslin, M. J., ...Renger, J. J. (2011). Promotion of sleep by suvorexant-a novel dual orexin receptorantagonist. Journal of Neurogenetics, 25(1–2), 52–61.

Winters, W. D., Ferrar-Allado, T., Guzman-Flores, C., & Alcaraz, M. (1972). The catalepticstate induced by ketamine: A review of the neuropharmacology of anesthesia.Neuropharmacology, 11(3), 303–315.

Zijlmans, M., Jacobs, J., Zelmann, R., Dubeau, F., & Gotman, J. (2009). High-frequency oscil-lations mirror disease activity in patients with epilepsy. Neurology, 72, 979–986.