IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

Embed Size (px)

Citation preview

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    1/20

    Fax +41 61 306 12 34

    E-Mail [email protected]

    Review

    Neuropsychobiology 2012;66:201220

    DOI: 10.1159/000343478

    Guidelines for the Recording and Evaluationof Pharmaco-EEG Data in Man:The International Pharmaco-EEG Society (IPEG)

    Marc Joberta Frederick J. Wilsonb G S.F. Ruigtc Martin Brunovskyd Leslie S. Prichepe

    Wilhelmus H.I.M. Drinkenburgf The IPEG Pharmaco-EEG Guidelines Committee

    aInternational Pharmaco-EEG Society, Berlin, Germany; bPharmatherapeutics Precision Medicine, Pfizer Ltd,

    Sandwich, UK; cClinical Consultancy for Neuroscience Drug Development, Oss, The Netherlands; dPsychiatric Center, Department

    of Psychiatry and Medical Psychology, Charles University, Prague, Czech Republic; eBrain Research Laboratories, Department ofPsychiatry, University School of Medicine, New York, N.Y., USA; fNeuroscience Discovery, Janssen Research and Development,

    Pharmaceutical Companies of Johnson and Johnson, Beerse, Belgium

    data in man. Since the publication of the first pharmaco-EEG

    guidelines in 1982, technical and data processing methods

    have advanced steadily, thus enhancing data quality and ex-

    panding the palette of tools available to investigate the ac-

    tion of drugs on the central nervous system (CNS), determine

    the pharmacokinetic and pharmacodynamic properties of

    novel therapeutics and evaluate the CNS penetration or tox-icity of compounds. However, a review of the literature re-

    veals inconsistent operating procedures from one study to

    another. While this fact does not invalidate results per se, the

    lack of standardisation constitutes a regrettable shortcom-

    ing, especially in the context of drug development pro-

    grammes. Moreover, this shortcoming hampers reliable

    Key Words

    -Activity Biomarkers Central nervous system

    Cordance Drug development Electroencephalography

    Guidelines Pharmacodynamics Pharmaco-EEG

    Pharmacokinetics Quantitative

    electroencephalography Source localisation

    Translatable EEG biomarkers

    Abstract

    The International Pharmaco-EEG Society (IPEG) presents

    updated guidelines summarising the requirements for the

    recording and computerised evaluation of pharmaco-EEG

    Received: May 14, 2012

    Accepted after revision: September 17, 2012

    Published online: October 12, 2012

    Marc JobertE-Mail publications @ ipeg-society.org

    2012 S. Karger AG, Basel0302282X/12/06640201$38.00/0

    Accessible online at:www.karger.com/nps

    Co-authors (in alphabetical order):

    Claudio Babiloni:Department of Biomedical Sciences, University

    of Foggia, Foggia, Italy; Peter H. Boeijinga: FORENAP Research and

    Development, Rouffach, France; Dominic H. ffytche:Institute of Psy-chiatry, Kings College London, London, UK; Jon Freeman:Brooklyn

    Strategic Consulting, New York, N.Y., USA; Joop M.A. van Gerven:

    Center for Human Drug Research, Leiden, The Netherlands; Koichi

    Hirata: Department of Neurology, Dokkyo Medical University, To-

    chigi, Japan; Ulrich Hegerl:Department of Psychiatry, University of

    Leipzig, Leipzig, Germany; Toshihiko Kinoshita:Department of Psy-

    chiatry, Kansai Medical University, Moriguchi, Osaka, Japan; Verner

    J. Knott: Institute of Mental Health Research, University of Ottawa,

    Ottawa, Ont., Canada; Fernando H. Lopes Da Silva:Center of Neu-

    roscience, Swammerdam Institute for Life Sciences, Amsterdam, The

    Netherlands; Milos Matousek: Department of Clinical Neurophysi-

    ology, Gteborg, Sweden;Armida Mucci:Department of Psychiatry,

    University of Naples SUN, Naples, Italy;Judith F. Nottage:Institute ofPsychiatry, Kings College London, London, UK; Sebastian Olbrich:

    Department of Psychiatry, University of Leipzig, Leipzig, Germany;

    Bernd Saletu:Department of Psychiatry and Psychotherapy, Medi-

    cal University of Vienna, Vienna, Austria; Andrej Stancak:Depart-

    ment of Experimental Psychology, Institute of Psychology, Health

    and Society, University of Liverpool, Liverpool, UK; Werner K. Strik:

    University Hospital for Clinical Psychiatry, Bern, Switzerland; Richard

    G. Wise:Cardiff University Brain Research Imaging Centre, School of

    Psychology, Cardiff University, Cardiff, UK.

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    2/20

    Jobert et al.Neuropsychobiology 2012;66:201220202

    comparisons between outcomes of studies from different

    laboratories and hence also prevents pooling of data which

    is a requirement for sufficiently powering the validation of

    novel analytical algorithms and EEG-based biomarkers. The

    present updated guidelines reflect the consensus of a glob-

    al panel of EEG experts and are intended to assist investiga-

    tors using pharmaco-EEG in clinical research, by providingclear and concise recommendations and thereby enabling

    standardisation of methodology and facilitating compara-

    bility of data across laboratories.

    Copyright 2012 S. Karger AG, Basel

    Introduction

    Pharmaco-electroencephalography (pharmaco-EEG)concerns the description and the quantitative analysis ofthe effects of substances on the central nervous system

    (CNS) by means of neurophysiological and electrophysi-ological methods used within the framework of clinicaland experimental pharmacology, neurotoxicology, thera-peutic research and associated disciplines. For the re-mainder of this article, the acronym pharmaco-EEGstrictly refers to human quantitative electroencephalog-raphy (QEEG) in the context of drug testing. Separateguidelines for pharmaco-sleep studies in man are inpreparation for publication by the International Pharma-co-EEG Society (IPEG). Evoked potentials (EPs)/event-related potentials (ERPs), in which the time-locked EEGsignal resulting from a specific event is examined, repre-

    sent another area of study and will be the subject of ded-icated guidelines.

    In the 80s and early 90s, several guidelines were pub-lished with the goal to standardise the acquisition andprocessing of data collected in pharmaco-EEG studies[13] or to provide procedural recommendations for therecording and quantitative analysis of EEG activity in re-search contexts [4].

    The initial pharmaco-EEG guidelines were comple-mented in 1995 by another guideline paper providing theframework to build standard operating procedures(SOPs), identifying the key components and minimum

    requirements for data acquisition, amplification and fil-tering, the validation of hardware and software, artefacttreatment and fast Fourier analysis [5]. This frameworkconstituted the basis for setting up laboratory-specificSOPs. This attempt was driven by the mandatory need forcompliance to good clinical practice, an internationalquality standard launched by the International Confer-ence on Harmonisation (ICH), an international body de-

    fining standards that governments can transpose intoregulations for the registration of pharmaceuticals andfor the conduct of clinical trials involving human sub-jects. Good clinical practice requires SOPs to be availableto document all methods and procedures in use duringdrug development.

    In parallel, several organisations published recom-mendations and guidelines for the use of EEG in variousclinical fields [68] in an effort to improve standardisa-tion and facilitate the proper utilisation of the techniquein clinical practice.

    EEG is a non-invasive method which directly revealsthe spontaneous synchronised postsynaptic neuronalactivity of the human cortex with high temporal resolu-tion. While EEG parameters are among the biomarkerswith the highest heritability [912], they show at thesame time a very high sensitivity to changes in both en-

    vironment and internal states (state-modulated traits).

    The sensitivity to such factors, which are extraneous tothe objective of many studies, means that a high degreeof quality control and detailed SOPs are required in or-der to decrease the effect of confounders in the analysisof the data.

    While pharmaco-EEG has demonstrated its value inthe development of CNS-active compounds in many in-stances, and while validated quantitative methods havebeen available for a long time to study the effects of drugson brain functions in patients and volunteers [13], thereis still reluctance to apply this method in large-scale clin-ical trials or for pivotal drug studies. There are a number

    of reasons contributing to this situation:(1) While there is evidence indicating the putative utility

    and validity of EEG as a biomarker relevant to a rangeof drug classes covering several therapeutic indica-tions, it has not yet been generally accepted as such.Further, the translatability of pharmaco-EEG signa-tures is not universal across the spectrum of CNS-ac-tive drugs, but depends on the pharmacological mech-anism and the preclinical species used. Hence its useas a translational biomarker for the preclinical screen-ing of compounds and the development of new drugsrequires careful interpretation [14].

    (2) Despite the fact that pharmaco-EEG has been used inresearch laboratories for several decades now [15, 16],operating procedures have not yet been standardisedto an extent facilitating reliable comparison of datasetsand results across units, making it difficult (or evenimpossible) to share datasets between sites or to poolresults from different clinical trials.

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    3/20

    IPEG Guidelines for Pharmaco-EEG Data Neuropsychobiology 2012;66:201220 203

    (3) This lack of standardisation constitutes a difficult ob-stacle for the design and interpretation of clinical trialsdue to the difficulties in comparing results across theliterature.

    (4) The large inter-individual variability observed in EEGrecords, some of which may be attributable to the lack

    of standardisation, can be compensated by increasingthe sample size, provided that the behavioural andawareness state of the subjects is tightly controlled.However, trial costs quickly become prohibitive as thesample size increases and hence improvements instandardisation which enable sample size reductionswill make routine application more attractive. Despitethis, EEG is still one of the cheapest of the variousmethods that can be used as a window into the brainsactivity, and also has the advantage that it can feasiblybe integrated into first-in-human studies.

    (5) For a long time, the amount of data generated by re-

    cording EEG signals from multiple electrodes quicklyoverwhelmed the storage capacity of computers andthe processing techniques were constrained by CPUpower. These limitations have now disappeared as aconsequence of vastly increased computing power andstorage capacities.In this context, one of the crucial steps is to enhance

    the standardisation of the operating procedures, not onlyto improve the ability to compare EEG data and resultsgenerated in different laboratories by reducing variancebut also to facilitate the creation of a centralised reposi-tory where a large number of records could be stored and

    shared. Such a repository would enable the following en-deavours:(1) Constitute reference datasets (i.e. both the raw EEG

    signals recorded against a unipolar reference and thederived parameters) obtained under standardisedconditions from different studies using various drugs(with emphasis on reference drugs and including pla-cebo) and study populations (healthy volunteers andvarious patient populations) under standardised be-havioural conditions, enabling comparative analyses.

    (2) Identify EEG parameters and properties that could beexploited as potential (translatable) biomarkers and

    quantify their validity on large populations.(3) Facilitate the transition of novel compounds from pre-

    clinical to clinical research and development pro-grams by enabling the early comparison of results ob-tained in preclinical screening and early clinical ex-periments, thereby improving the decision-makingprocess as well as de-risking and accelerating the de-velopment of new CNS-active compounds.

    Pharmaco-EEG Studies

    FundamentalsPharmaco-EEG studies are applicable when there are

    indications that a substance may have an effect on theCNS at therapeutic or pharmacologically relevant dose

    levels. Pharmaco-EEG studies are indicated in all phasesof clinical research and may answer questions in clinicalpharmacology as well as in therapeutic research. It is theconsistency and nature of the empirical observations cap-tured by the pharmaco-EEG method, particularly in therealm of clinical psychopharmacology, which have estab-lished this technique as a suitable means for a completequantitative description of the effect of a drug on thefunction of the brain in humans [17, 18]. Pharmaco-EEGdata can be exploited to classify psychotropic substances[19, 20] and to assess drug-drug interactions or to moni-tor side effects and toxicity [21]. The use of quantitative

    methods for data reduction and statistical analysis pro-vides quantitative descriptions of the direct and indirecteffects of active compounds on brain functions [22] hencegenerating pharmacodynamic (PD) outcome measureswhich, along with separate pharmacokinetic (PK) data(e.g. drug concentration from blood samples), can beused to study the PK-PD relationship [2325].

    Pharmaco-EEG studies need to be designed in accor-dance with the objectives of the study (including placebocontrol and/or reference drug and test-retest designwhen indicated, and always keeping the behavioural andawareness state of the subjects under well-controlled

    standardised conditions) and require appropriate statis-tical methods adapted to the study design and the inves-tigated hypotheses. Dependent upon the study objec-tives, either patients and/or normal volunteers may beexamined. Although a given psychotropic drug tends toproduce similar and replicable EEG profile changes indifferent populations at the group level, per-subject dif-ferences in EEG response profiles have also been shownto be a useful indicator of the biological variability andtherapeutic potency of a compound. Less potent psycho-tropic drugs often exhibit higher biological variability(as seen in individual EEG response profiles), while more

    therapeutically potent compounds (i.e. with low biologi-cal variability) generally produce very similar EEG pro-file changes in the majority of subjects tested [26]. In pa-tient populations, clinical response to drugs has shownto be a source of variation with different patterns ob-served in responders versus non-responders [27, 28].This illustrates the potential utility of EEG for patientselection in clinical trials or as a companion diagnostic

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    4/20

    Jobert et al.Neuropsychobiology 2012;66:201220204

    to certain drug therapies, thus facilitating a personalisedmedicine approach.

    All clinical trial programmes must follow the ICHguidelines aimed at ensuring that good quality, safe andeffective medicines are developed and registered in themost efficient and cost-effective manner. These activitiesare pursued in the interest of the consumer and publichealth, to prevent unnecessary duplication of clinical tri-als in humans and to minimize the use of animal testing

    without compromising the regulatory obligations of safe-ty and effectiveness. The ICH guidelines cover a broadrange of activities related to drug development [29, 30].

    In addition, the responsible investigator should: (a) beacquainted with the literature and techniques of EEGdata processing which are the basis for the interpretationof the results; (b) understand the range of normal andabnormal variability of the neurophysiological methodsused, and (c) be responsible for the planning, organisa-tion and execution of the study.

    Subjects

    Due to the broad spectrum of neurophysiological do-mains where pharmaco-EEG studies are applicable, it isnot possible to provide an all-inclusive standard for sub-ject-related data which should be collected in pharmaco-EEG trials. The type of subject data to be collected maydepend upon the objectives, targeted population, pathol-ogy (by patients) and other attributes of a specific trial.

    The following factors should be considered when com-piling inclusion and exclusion criteria for a particularstudy and documented routinely: demographic data (age,gender, handedness and socioeconomic status); medicalhistory [state of health, prior illnesses, presence of any

    metabolic syndrome, hyperglycaemia or thyroid disor-der, use of drugs (including medication and recreationalsubstances, particularly cannabis), sleep quality and be-haviour, and EEG characteristics]; psychiatric history(DSM or ICD diagnoses), and use of tobacco, coffee, tea,energy drinks and alcohol (before and during the days ofexamination). If relevant, more personal characteristicsare to be filed: emotional lability, neuroticism, extraver-

    sion/introversion; important psycho-physiological attri-butes such as emotional state (anxiety or fatigue), reac-tion to stress, bladder or bowel problems and menstrualcycle, for example. Where possible, standardised ratingscales should be used to quantify characteristics and sub-jective descriptors should be avoided.

    Table 1 summarises the set of subject metadata to berecorded in pharmaco-EEG trials.

    Given the profound effect of nicotine, caffeine and al-

    cohol consumption on the EEG signal, it is generally rec-ommended to restrict the use of these products in clinicalEEG trials, for instance by excluding subjects if they meetany of the following conditions within the previous 6months: History of regular alcohol consumption exceeding 23

    units/day for females and 34 units/day for males [31] History of regular use of tobacco or nicotine-contain-

    ing products exceeding the equivalent of 5 cigarettesper day

    History of regular consumption of caffeine exceedingthe equivalent of 4 cups of coffee per day, a level that

    approximates health-related criteria [32]In addition, subjects should refrain from alcohol and

    caffeine for at least 24 h, and from tobacco or nicotineproducts for at least 4 h (preferably 8 h) prior to an EEGrecording.

    Stricter exclusion criteria or restrictions may be ap-plied on a study-by-study basis where required.

    Phenotyping and/or genotyping of the participants isacquiring increasing importance for safety and PK rea-sons; since drugs can be primarily metabolized throughspecific cytochrome P450 (CYP) isoenzymes, metabolicstatus could be defined accordingly in order to avoid po-

    tential accumulation/fast elimination during the wash-out phase, for example. If a drug is known to be subjectto a major genetic polymorphism, studies could be per-formed in panels of participants of known phenotype orgenotype for the polymorphism in question.

    In addition, genotyping is becoming relevant for theEEG itself: it has been demonstrated that a low/high nat-ural rhythm can influence the interpretation of the

    Demographics Age, gender, handedness

    Medical history State of health, prior or current illnesses, psychiatric history

    Screening EEG Abnormalities, high/low power

    Medication status Current use of drugs, use of drugs in the past 4 weeksUse of alcohol, tobacco, illicit drugs

    Table 1.Minimum set of subjectmetadata to be filed in pharmaco-EEGtrials

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    5/20

    IPEG Guidelines for Pharmaco-EEG Data Neuropsychobiology 2012;66:201220 205

    EEG response to drugs [33], and studies of twins concludethat the high inter-individual variability in power andpeak frequency is largely genetically determined [912].Today, the best candidate is a gene encoding catechol-O-methyltransferase. According to a recent study [34], theVal158Met polymorphism of the enzyme is highly predic-

    tive of individual activity, with the Val allele carriersdisplaying lower peak frequency and lower activitycompared to the Met allele carriers. If this variability islikely to have an effect on a particular study, then suitablesubjects could be selected either by genotyping or byscreening for low/high EEG rhythm. Alternatively, ge-notyping data could be collected for post hoc analysis.

    Pharmaco-EEG Data Acquisition

    Digital Recording

    Digitising is the conversion of an analogue (continu-ous) signal into a digital (discrete) representation (i.e. asequence of numbers). Analogue-to-digital (A/D) con-verters usually have a resolution of 16 bits, meaning thatthe analogue amplitude of each discrete point is repre-sented by a value ranging from 0 to 65,535 (0 to 216 1).The sampling rate (TS) corresponds to the time intervalbetween two subsequent points and determines the reso-lution in time. The sampling frequency (FS) expresses thenumber of samples digitised per second and is the recip-rocal of the sampling rate (FS= 1/TS). With a samplingfrequency of 500 Hz, the resolution in time (s) corre-

    sponds to the sampling rate and is given by the reciprocalof the sampling frequency, i.e. 2 ms.

    From a theoretical point of view, the sampling fre-quency must be at least twice the highest frequency pres-ent in the signal to be digitised (Nyquist-Shannon sam-pling theorem). Conversely, frequency components high-er than half the sampling frequency (also called Nyquistfrequency, FN= FS/2) must be removed using analoguefilters before digitising to avoid aliasing effects. Errorsintroduced in the digitised signal by aliasing cannot bedetected and corrected afterwards. Because of imperfec-tions in the analogue filters, the sampling frequency is in

    practice at least fourfold the analogue filter (anti-alias-ing) cut-off frequency.

    From a practical point of view, the following appliesfor pharmaco-EEG studies: Sampling frequency must beat least 500 Hz (i.e. 500 samples per second) and be a mul-tiple of 50 or 64 [e.g. 500 or 512 Hz (currently in neuro-cognitive studies and in epilepsy, EEGs are being record-ed with a wider frequency band to allow the study of high

    band components or, more generally, high frequencyoscillations; in this case, sampling has to be at rates 12kHz. This is discussed in the section on High-Frequency() EEG Activity below)]. The A/D converter must have adigital resolution of at least 16 bits and resolve changes inthe EEG below 0.2V. Prior to sampling, an anti-aliasing

    low-pass filter set at 70 Hz (with a roll-off of at least 12dB/octave) must be used and the high-pass filter shouldbe set below 0.5 Hz (time constant 12 s; recommended0.01 Hz). Ideally, digital recordings should be made withminimal filtering (as above) and additional filters appliedpost hoc. This enables the effect of the filtering step to beevaluated. In particular, the use of a notch filter (50 or 60Hz) during recording should be avoided as it can poten-tially disguise an electrode problem, while the mainsnoise can be eliminated off-line at the data processingstage.

    The electrode impedance (or resistance) should con-

    ventionally be maintained below 5 k. However, modernamplifiers with high internal resistances are able to re-cord at higher scalp impedances (e.g. without using con-ductive gel). Similarly, MRI compatible EEG caps haveinbuilt safety resistors that set a minimum impedance ofaround 10 k. In these cases, higher impedance is accept-able, but in all cases it is important to balance impedanceacross electrode sites. The preamplifier input impedancemust be over 100 M. As the rejection of cross-talk be-tween channels is important for coherence or other mea-sures of relationships between electrodes, a cross-talk re-jection of at least 90 dB is required and better is recom-

    mended.Comprehensible overviews on technical aspects relat-

    ed to the digital recording of EEG signals have been pub-lished elsewhere and provide additional insight on spe-cific details [3537].

    CalibrationRecording accuracy (how far the sample varies from

    the true signal value) is dependent upon system calibra-tion. The calibration procedure is aimed at testing theperformance of the entire hardware and must be carriedout before each measurement. Calibration is also essen-

    tial to achieve a reference potential of known voltageagainst the absolute amplitude of the recorded signals. Topick up possible time-dependent fluctuations of ampli-fiers, due to thermic effects for example, it is recommend-ed to re-check the calibration at the end of each measure-ment session.

    Nowadays, most EEG machines have internal hard-ware calibration and the procedure may be carried out

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    6/20

    Jobert et al.Neuropsychobiology 2012;66:201220206

    automatically. Verification is made that the same inputsignals (sine waves with known amplitude and frequen-cy) applied to all channels are present with the same am-plitude at the output of the amplifiers and are subse-quently correctly transmitted to the A/D converter. If in-ternal calibration is not available, then an external device

    should be used to generate stable test waves that are re-layed through the electrode sockets.

    Ideally, standardised calibration procedures shouldinclude sine waves of stable amplitude (e.g. 100V) at twodifferent frequency ranges (e.g. at 1 and 10 Hz) whetherinternally generated or from an external source. Verifica-tion of the calibration signal should be performed withdigital callipers utilising as long a duration of the calibra-tion signal as possible (e.g. 1 min per frequency). Al-though no definitive data currently exist on the amountof tolerable deviation for pharmaco-EEG signals, a max-imum acceptable variance of82% in signal amplitude at

    both frequencies is recommended. Between-channel dif-ferences suggest an amplifier imbalance and channelswith182% variance should be eliminated from analysis.

    Electrode PositionsWhile demonstration has been made in the past that

    it is possible to trace the effects of specific compoundswith a limited number of electrodes (e.g. using the bipolarfronto-central and occipito-temporal derivations, Fz-Czand Oz-T6), it is no-longer state-of-the-art and inappro-priate for advanced investigations. Instead, a configura-tion of 21 electrodes placed according to the internation-

    al 1020 system [38] is the recommended minimal elec-trode configuration. If additional EEG electrodes areused (e.g. for mapping or localisation studies), then theinternational extended 1020 electrode placement sys-tem (also known as 10% system [3941]) should beutilised. The nomenclature of the electrodes must followthese standards.

    Many laboratories use electrode caps (as opposed tomeasuring individual electrode positions) for ease of ap-plication of dense electrode arrays. However, cap elec-trode positions only approximate the 1020 locations andare rarely measured accurately. It is important to ensure

    that the electrode location does not deviate from the stan-dard when using caps.

    As a matter of principle, recording against one refer-ence electrode is recommended (e.g. against Cz, A1andA2) or the arithmetic mean of A1and A2[(A1+ A2)/2] (i.e.a mathematically generated linked-ear or linked-mas-toid reference). Without resistors between them, twolinked electrodes (e.g. A1with A2) connect two brain re-

    gions, thereby changing the electrical potential distribu-tion over the scalp. Hence, although the resulting distor-tion of potential may be negligible in many or even mostcases [42], physically linking the electrodes is not recom-mended. A linked reference may also be problematic ifthe impedance of one or other electrode varies differen-

    tially during a recording (for example if one of the elec-trodes becomes dislodged). The data must be stored in aformat permitting conversion from the recording refer-ence to any other reference (common average reference,current source derivation or other channels as reference,for example). Active electrode systems can be used inpharmaco-EEG studies provided that active electrodeswould not distort any frequency components in the fre-quency range 0.0540 Hz.

    In addition, EOG (electrooculography) must be re-corded (vertical and horizontal, bipolar) to aid in identi-fying artefacts and to correct off line the interference cap-

    tured in the EEG using appropriate data processing algo-rithms. The recording of both ECG (electrocardiography)for assessment of the activity of the autonomous nervoussystem and EMG (electromyography) as additional mark-er for vigilance stages or artefacts is recommended. Therecording of other vital parameters, such as blood pres-sure and respiration, is optional.

    Environmental ConditionsThere are many environmental factors affecting the

    function and activity of the CNS and, as a result, also af-fecting neurophysiologic readouts of brain activity. It is

    therefore necessary to control these factors to the bestpossible extent. If deviations from normal, pre-existingor pre-defined conditions are observed, then these shouldbe recorded as metadata. In clinical trials, it is manda-tory to document the degree to which these factors havebeen used as inclusion and exclusion criteria.

    Adaptation. EEG measurements are subject and sensi-tive to adaptation effects. Consequently, at least one pre-examination EEG recording (apart from the screeningEEG) should ideally be performed on a separate day tocarry out one (or several) blank or habituationmeasurement(s). Such a procedure allows the subject to

    become familiar with the environment, apparatus and re-cording protocol. When this additional subject visit is notfeasible, then a period of acclimation should be includedin the protocol on the study day itself before the actualrecording session is started.

    Room. The recording should occur in a separate,sound-attenuated room with constant light (approxi-mately 40 lx), regulated temperature (2023 C/6873F)

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    7/20

    IPEG Guidelines for Pharmaco-EEG Data Neuropsychobiology 2012;66:201220 207

    and normal humidity conditions. Intermittent disturb-ing events must be avoided. Random and undefined in-teractions between the subject and the staff during therecording (as for changing the electrode placement orposture, or giving additional instructions or stimulation)should be limited and documented. Typically, clinical

    neurophysiology studies are carried out with the subjectsitting in a half-reclined position to reduce neck muscletension and with constant dimmed light (approximately40 lx) and this is recommended in many cases. However,these conditions are not amenable to the presentation ofa task and so resting data acquired in this way will not besuitable for comparison with subsequent task-related re-cordings forming part of the same study. Hence in studiesinvolving both resting and task-related recordings, it isrecommended that resting recordings be made in a situ-ation that exactly replicates the recording conditions tobe used when engaged in tasks (i.e. the subjects should be

    comfortably seated in a suitably supportive high-backedchair in an upright position). The ambient light levelshould be maintained at a similar level during resting andtask-related recordings.

    Time of Recording. Recording periods should be com-pleted at the same time of the day and under the sameconditions. If possible, having documentation of a sub-jects wake and sleep history (e.g. around 1 week of con-sistent bed- and rise-time) is advisable to assist in recog-nising circadian influences on the EEG (subjects withhighly disrupted sleep should be carefully evaluated andpotentially excluded from the study). Whenever possible,

    the recording should preferably occur in the morning(between 9: 00 and 13: 00 h) to avoid interference withmeal times and postprandial vigilance f luctuations. Thisaspect is to be taken into consideration for the design ofstudies with repeated measurements following drug in-take (e.g. at baseline and 1, 2, 4 and 8 h af ter medication).Also the type of food (breakfast and lunch) must be care-fully selected.

    Recording ConditionsFor the purpose of standardisation and to ensure that

    results obtained in one laboratory can be compared with

    results obtained in another, pharmaco-EEG studiesshould be recorded under one or more of the followingconditions:

    (1) Vigilance-controlled EEG for 5 min (RT).Duringthis recording session, the subjects must have their eyesopen and the vigilance level should be controlled by asimple continuous performance task (e.g. creating a storyout of given words or solving simple arithmetic prob-

    lems), which incorporates a fixation point to minimiseeye movement artefacts. The purpose is to stabilise thevigilance to a relatively narrowly defined level by meansof the continuous mental exercise. The EEG should bemonitored online by an EEG technician and short exter-nal interventions for vigilance stimulation are permissi-

    ble if a decrease in vigilance is observed, for instance incase of intrusion of slow-wave activity in the EEG. How-ever, the EEG segments in which the interventions oc-curred are labelled as artefacts and are excluded from theevaluation.

    (2) Resting EEG with closed eyes for 5 to 15 min (RSc).During this session, the EEG is recorded with closed eyesand fluctuations of vigilance are permitted without anyrestriction, the purpose of this recording condition beingto examine the variations in vigilance over time. It is es-sential that the instructions should be explicit, such as Sitquietly and keep your eyes closed; there is no task to com-

    plete, just relax! Limited external interventions shouldtake place during the session and used only when neces-sary to help maintain consistency of state (e.g. eyes open-ing). Drowsiness EEG patterns or falling asleep are not areason to intervene. While sessions of 5 min yielding aminimum of 2-min artefact-free signal are sufficient toquantify EEG activity and demonstrate pharmacologicaleffects on spectral parameters [43], the recording may beextended to 15 min to assess the regulation of vigilance(CNS arousal) and wakefulness, and to quantify drug-induced changes in these parameters (e.g. sleepiness)[44].

    (3) Resting EEG with open and closed eyes for 5 min ineach eye state (RSco).Recordings of this type employ al-ternate periods with eyes open and closed of 1-min dura-tion each in response to commands from the technician.Subjects should face a featureless wall in order to stan-dardise the visual environment and reduce the effect ofeye movements on the EEG during periods with openeyes. No additional vigilance control procedure should beused; in particular, fixation is not recommended as thisin itself constitutes a task. Data in each of the two statesshould be separated prior to processing and the EEG ac-tivity analysed separately for segments with open and

    closed eyes.

    Important Comments: The task to be used during the vigilance-controlled

    condition has deliberately not been tightly defined inorder to encompass the range of currently acceptedpractice. One particular variation is to use a visualperformance task undertaken at a computer terminal,

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    8/20

    Jobert et al.Neuropsychobiology 2012;66:201220208

    which has the benefit of also providing a measure ofperformance whereby epochs surrounding stimuluspresentation and motor response or outside presetlimits of normal reaction time can be excluded fromthe analysis. There are also other accepted proceduresfor maintaining vigilance during EEG recording, in-

    cluding the use of a device which is sensitive to musclerelaxation. However, it is important to note that theresults cannot be readily compared when differenttasks or vigilance control methods are used as system-atic differences in both the topography and frequencycomposition of the resulting EEG signals are to be ex-pected. Hence, the method used must always be clear-ly described alongside the results.

    Continuous video recording synchronised with theEEG is recommended.

    It may not be possible to fully adhere to the standardprocedures in cases where patients are uncooperative

    or unable to follow instructions (e.g. psychotic or se-verely demented patients). Any deviations from thestandard must be clearly stated alongside the presenta-tion of the results.

    EEG recording should always take place prior to ad-ditional testing (e.g. cognitive paradigms), since vigi-lance might be decreased after exhausting or time-consuming tests.

    Since the brain activity indicated by the EEG dependsstrongly on the present level of vigilance [45], care hasto be taken that drug effects on vigilance regulation(e.g. more rapid decline to lower vigilance stages with

    sedating drugs) are separated from drug effects onEEG activity within the same vigilance level (e.g. in-crease in frequency).

    With measures such as connectivity, phase and coher-ence, there is evidence that more data are necessary toconverge to stable, reliable estimates, and hence therecording duration for any of the conditions may beextended if required to meet the objectives of the study.

    ArtefactsArtefact identification and elimination is crucial for

    the proper quantitative analysis of EEG records. With op-

    timal recording conditions, artefacts are only accidentaland appear infrequently. Artefacts can have various phys-iological origins (ocular, cardiac, muscular, behavioural,sweating and respiratory) and can be identified and/oreliminated either on line during the recording or off line.Special care should be taken in those studies where high-er frequency bands (i.e. and ) are of interest since theEEG signals may be contaminated by muscle activity.

    Automated EOG artefact rejection is possible usingappropriate algorithms [4648]. However, when comput-erised algorithms are used, a semi-automatic procedurethat includes additional visual inspection is recommend-ed. Even new approaches such as the independent com-ponent analysis (ICA) used for the detection of eye move-

    ment or muscle artefacts [49] should be applied with cau-tion. The risk of excluding components with mixedneurophysiological information of brain activity and ar-tefacts should be minimised by careful inspection of top-ographic maps of the components and by using an appro-priate number of components (equal to the number ofchannels of the input matrix).

    Should the investigator doubt the validity of the pro-cedure either because of the large percentage of EEG seg-ments containing artefacts or because the kind of arte-facts could be confused with the treatment effect, then acomparative biometrical evaluation and assessment of

    the artefact-free and complete data should follow.

    Minimum RequirementsTable 2 summarises the minimum requirements for

    the recording of pharmaco-EEG studies.

    Pharmaco-EEG Data Processing

    Representation in the Time DomainIn the time domain, the variations in potential after

    amplification are displayed as a function of time and sig-

    nals are usually denoted by a function s(t) [or s(k TS)(with k = 1N) in its digital form]. The representation inthe time domain is used for the visual inspection of EEGcurves and for the evaluation of EEG activity resultingfrom external sensory stimulation for which the positionin time is relative to the time point at which a stimulusoccurs. Thus, time is considered as a variable of the ob-served phenomenon. The detection of patterns or tran-sient activities in the EEG usually relies on processingalgorithms operating in the time domain.

    Representation in the Frequency Domain

    The transformation of a signal s(t) into the frequencydomain using the fast Fourier transformation (FFT) im-plicitly assumes that s(t) can be split up as a finite sum ofweighted sinusoidal waveforms [denoted as s(f)]. Thenumber of sinusoidal waveforms is dependent upon thewindow size (i.e. the number of points of the input signal)subjected to FFT. The resulting graphical representationdisplays the spectral characteristics of s(t), which is then

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    9/20

    IPEG Guidelines for Pharmaco-EEG Data Neuropsychobiology 2012;66:201220 209

    Table 2.Minimum requirements for the recording of pharmaco-EEG studies

    EEG recordingequipment

    Sampling rate 500 Hz

    A/D conversion 16 bits

    High-pass filtering 0.5 Hz (0.01 Hz recommended)

    Low-pass filtering 70 Hz (roll-off of at least 12 dB/octave)

    Notch filter Usage avoided; otherwise 50 or 60 Hz (dependent on the power supply frequency)Pre-amplifier impedance 100 Mat 50 Hz

    Common mode rejection 90 dB

    Electrodes Electrode impedance Balanced impedance across all electrode sites

    Number and placement At least 21 electrodes placed according to the 1020 system or the extended1020 system (10% system) in case >21 electrodes are used

    Type Ag/AgCl or equivalent in terms of electrode drift and DC resistance

    Montage Monopolar against a common reference

    Reference Should be modifiable post hoc, (Cz, A1, A2, average mastoids)

    Ground AFzEOG Vertical and horizontal for artefact identification

    ECG Recommended

    EMG RecommendedExperimentaldesign andconditions

    Adaptation It is recommended to make the subject familiar with the recording conditions andprocedures during a separate recording session

    Recording environment Sound-attenuated roomConstant dimmed light (approximately 40 lx)or light level defined by the computer monitor used for task presentationConstant room temperature: 2023C (6873F)Any major disturbances should be loggedSubject in a (semi-)reclined comfortable position or in an upright position facing acomputer monitor (for studies that also include a task)

    Design Double-blind placebo-controlled cross-over design is recommended for acute studiesin healthy subjectsFor multiple dose and patient studies, the design should be adapted according to theobjectives

    Recording time points Baseline and a number of post-drug recording time points to be driven by drug PK; atleast one time point around Tmaxplus at least 3 time points covering the decline in thePK curve (usually multiples e.g. 1, 2, 4 and 8 h)If Thalf12 h, then a 24-hour overnight time point should be considered

    Time of day Preferably in the morningCross-over repeat tests should be done at the same clock time and under the sameconditions

    Recording conditions RT (5 min vigilance controlled, eyes open)RSc(5- to 15-min resting condition, eyes closed)RSco(10-min resting with alternate eyes open and eyes closed)

    Storageconditions

    Local storage The proprietary format of each EEG recording equipment

    Export/import format European data format plus [see ref. 50 for details]

    Signals Raw data without transformation

    Automatic artefact rejection is optional

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    10/20

    Jobert et al.Neuropsychobiology 2012;66:201220210

    depicted by peaks in the frequency domain. This kind ofdisplay is generally used for the evaluation of spontane-ous EEG activity for which the position in time of eventshas no direct relevance.

    The FFT (as an orthogonal transformation) is essen-tially a mathematical operation performed on time seriesdata which does not alter the information content of thesignal. Neither is any assumption made regarding the na-ture of the data nor any interpretation implied. Withinthe limitations of computational accuracy, the full revers-ibility of the transformation is implicit and given only as

    long as numbers are retained in their complex form andnot averaged.

    The resolution in the frequency domain depends uponthe sampling frequency (FS) and the number (N) of sam-pling points (size of the signal window) subjected to FFT.This resolution (denoted here as F) is given by the ratio(F = FS/N). Accordingly, the longer the signal window(N), the better the resolution of the frequency content.

    When considering the results of an FFT applied to asignal window of N = 2,048 points (a number chosen be-cause it corresponds to 211and facilitates a rounded-offlength) with FS= 512 Hz, thenF = 0.25 Hz, which meansthat the frequency analysis can resolve 0.25 Hz (that isresolve 10.25 vs. 10.50 Hz directly). In this particular

    case, the signal window (called epoch) will have a lengthof 4 s.

    To reduce the broadband artefact, known as leakage,the signal window must be tapered toward zero at theirinitial and final data points (this tapering is usually doneusing a windowing function). When the FFT is appliedon sequential epochs, then discarding a proportion of thesignal through windowing can lead to differences inspectra depending on the starting point of the epoch se-ries. An alternative that results in a spectrum less sensi-tive to the starting point is to use partially overlappingepochs so that all data are represented.

    Although computer-based analysis of scalp-recordedsignals for pharmaco-EEG profiling has utilised a widerange of quantitative techniques, spectral analysis viaFFT is currently the most common method of choice forthe parameterisation of pharmaco-EEG studies.

    Non-StationarityMany signals, including EEG, are non-stationary,

    which means that they have a time-varying frequencyspectrum, although they can be considered locally sta-tionary over short segments in which the parameters ofinterest vary minimally.

    In practice, the choice of the segment length is a trade-off between frequency resolution (which suggests a longerepoch) and ensuring quasi-stationarity (which suggests ashorter epoch). For the pharmaco-EEG, epochs of 2- to10-second duration are used.

    Spectral AnalysisThe traditional parameterisation of pharmaco-EEG

    activity is largely based on spectral analysis. To this end,the recorded signals are divided into epochs (210 s)which are subjected to spectral analysis using FFT. Thistransformation in the frequency domain and subsequent

    computation of the power spectrum allows a first datareduction.

    The second step of data reduction consists in the extrac-tion of spectral parameters. The frequency range is subdi-vided into frequency bands, and the spectral performance(area under the curve) is computed for each of them (ex-pressed in V/Hz; square root of absolute power) or usinganother transformation (e.g. the natural logarithm of ab-

    Table 3.Frequency ranges for spectral analysis in pharmaco-EEGstudies

    Nomenclature Frequency rangeHz

    Units of theresults

    F 1.5 to

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    11/20

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    12/20

    Jobert et al.Neuropsychobiology 2012;66:201220212

    In the following and for the purpose of example, it willbe assumed that the EEG signals are recorded with a sam-

    pling frequency of 500 Hz (FS) and that the spectral pa-rameters are computed for epochs of 4-second length (i.e.2,000 samples), whereby the FFT are computed based on2,048 (211) points, with the individual overlapping epochsbeing tapered digitally toward zero voltage at their initialand final 24 data points. This way, 1 min of EEG record-ing will produce 15 power spectra, each 4-second epochproviding a set of spectral parameters (table 3).

    Considering an EEG signal recorded for 5 min underRT or under RScconditions, the 5-min recording is firstdivided into 75 epochs; artefact-marked epochs are elim-inated, and the sets of spectral parameters are calculated.

    Then, the strategy for data processing involves two steps(fig. 1):(1) For each spectral parameter, the activity over 5 min is

    quantified using the sample mean and its standard de-viation (the median and its quartiles or the trimeancan also be used). This procedure is straightforwardand provides for each measurement point and eachEEG channel a set of spectral values that can be com-pared with others. The main drawback of this ap-proach lies in the fact that it does not allow changesthat may occur during the 5-min recording to be cap-tured [43], e.g. as a result of a sedative effect. This prob-

    lem can be addressed by proceeding with a subsequentcomplementary assessment.

    (2) The spectral activity at the beginning and at the endof the recording are quantified separately and the re-sults compared. This assessment is particularly usefulto verify the stability of the recording condition (asexpected for RT) or to detect time-dependent changes(especially for RSc).

    Typically, the duration of the recording under RScisextended to up to around 15 min to assess the dynamicsof vigilance f luctuations. In this case, the vigilance regu-lation can be explored and vigilance levels assessed overthe whole duration of the recording session, from relaxedwakefulness to sleep onset [6870]. When focusing on

    spectral activities only, the recording can be decomposedinto segments of equal length (e.g. 3 min) and the spectralparameters quantified for each segment (fig. 2) using thesample mean and standard deviation or other suitable pa-rameters as described above, following the elimination ofepochs containing artefacts. Then, the results obtainedfor the segments at the beginning, the middle and the endof the recording can be compared to capture time-depen-dent changes.

    The processing of EEG signals recorded in the restingstate with alternate periods of RSco requires separatingthe epochs corresponding to the respective eye states

    (fig. 3). Off-line treatment of EOG (blink) events occur-ring in the open-eye segments is mandatory and requiresparticular attention (e.g. using ICA or similar filteringtechniques). Then, the spectral parameters can be quan-tified for each eye state separately using the same methodas for the RT and RScrecording conditions (fig. 1). Also,in this case, it is useful to verify the stability of the re-cording through assessment of any time-dependentchanges.

    In pharmacological trials aimed at assessing the effectof compounds on the CNS and on the EEG in particular,the study design as well as the selected recording condi-

    tions and related processing strategies will be dependentupon the profile of the compound under examination,whereby the hypotheses and targeted parameters will bedefined upfront. Exploratory evaluations, however, mayrequire more flexibility, especially when the potential ef-fects are unknown. When documented and justified, therecording duration and the strategy selected for data pro-cessing can be adjusted if specifically needed for a par-ticular study. However, such deviations may impede theability to compare results with other studies.

    Pharmaco-EEG Statistical Analysis

    The present guideline paper is not intended to discussthe statistical aspects of the design and analysis of phar-maco-EEG studies and especially of PD studies. To thisend, reference is made to the corresponding IPEG guide-lines [71], which provide a set of recommendations for theinvestigation of the PD characteristics of a compound

    10 2 3 4 5

    Recording duration (min)

    Fig. 1.Processing of 5 min EEG recorded under RT or RSccondi-tions. Spectral activity is analysed over 5 min (A). A comparisonof spectral activity for the segments at the beginning and the endof the recording (B) can be applied to capture time-dependentchanges.

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    13/20

    IPEG Guidelines for Pharmaco-EEG Data Neuropsychobiology 2012;66:201220 213

    (e.g. time or dose effects) in the early phases of clinicaldrug development.

    In summary, the key factors to consider during theplanning include, but are not limited to, the suitable typeof study design, the determination of the sample size andthe randomisation. In PD studies, the investigator mustaddress the problem of multiplicity of tests resulting

    from a profusion of parameters (i.e. inferential statisticalstatements) to which a statistical comparison is applied.Further, the computation of a large number of statisticaltests in a given sample may lead to false conclusions andmust be used careful ly [72]. In a paper published in 1987,Abt [73] distinguishes three levels of statistical approachbased on probability computations: exploratory, descrip-tive and confirmatory statistical analyses. The descrip-

    tive data analysis suggests inspection of changes by study-ing the pattern of descriptive p values assessed for allpairwise tests for differences in time or treatment. Thisapproach is particularly useful for the evaluation of topo-graphical changes [74]. Valid alternatives to this methodinclude randomisation and non-parametric permutationanalyses, which are used in brain imaging studies [75, 76],

    including EEG source imaging.Sources of multiplicity are several variables, time

    points and locations of measurement/observation, andcomparisons of more than two treatments or differentgalenic formulations. Moreover, pharmaco-EEG profilesmay also be examined in relation to behavioural changesassessed with rating scales, neuropsychological evalua-tions and computerised cognitive tests.

    10 2 3 4 5 6 7 8 9 10 11 12 13 14 15

    Recording duration (min)

    Fig. 2.Processing of 15 min EEG recorded under RSc. The variations in spectral activity can be analysed over15 min (C) or over consecutive segments (D). A comparison of spectral activity for segments of 3 min at thebeginning, middle and end of the recording can be applied to capture time-dependent changes (E).

    10 2 3 4 5 6 7 8 9 10

    Recording duration (min)

    eyes open

    eyes closed

    Fig. 3.Processing of 10 min EEG recorded under RS co. During the recording session, the subject alternates be-

    tween states with eyes open and closed every 60 s. The spectral activity is analysed over the 5 segments corre-sponding to (F) and (G) recording periods with eyes open and closed.

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    14/20

    Jobert et al.Neuropsychobiology 2012;66:201220214

    Whilst univariate models generally compare individ-ual variables of between-group or within-subject datas-ets, multivariate statistics assess multiple factors to creatediscriminants. Some methods utilise linear combina-tions of EEG measures with other clinical, cognitive andaffective ratings (neurometric analysis [77]). Methods

    based on EEG measures alone are less well developed butshow future promise [78], and this field is advancing rap-idly, driven mainly by the use of such techniques in func-tional MRI (fMRI).

    Finally, it should be noted that careful modelling andprecise correlation with independent parameters, such asPK factors, hormonal profile or metabolic status, are oftenneeded to determine the particular electrophysiologicalchange induced by a neuroactive drug, due to the inherentnon-specificity of the electrophysiological effects [79].

    Due to the complexity of most statistical analyses re-quired for the evaluation of pharmaco-EEG studies, the

    assistance of an experienced statistician is highly recom-mended.

    Use of Normative DatabasesWhile not always available, when appropriate norma-

    tive data exist [80], the data collected should be subjectedto comparisons (e.g. z-transformation) with a normativedata sample. The power spectrum of the EEG is extreme-ly stable and highly replicable, distinctive for each brainregion in a healthy, normally functioning individual, andis generally independent of cultural, ethnic or racial fac-tors. Therefore, this predictable, known power spectrum

    can be used as a normative baseline that specifies the ex-pected electrical activity of the healthy, normally func-tioning human brain across the human lifespan. Devia-tions from these norms can be described as standardscores, providing a reliable and objective metric for ab-normalities of brain electrical activity [8183].

    Pharmaco-EEG Advanced Topics

    EEG Source LocalisationThe ability to display the time course and localisation

    of brain activity based on extracranial measurements re-quires a mathematical solution to the inverse problem forthe computation of images of electric neuronal activitybased on the EEG signals recorded from the electrodes.In general, there is no unique solution to this problem.Inverse solutions critically depend on many a priori as-sumptions, including head models, solution spaces, regu-larisation parameters, the specif ic assumption on current

    density distribution and procedures used for statisticalmapping (like normalisation or log transformation) andstatistical thresholds. All these factors have a significantimpact on the results that may vary drastically dependingon the choices made and it is usually impossible to vali-date the correctness of the choices independently. Conse-

    quently, it is therefore absolutely necessary that all ofthese parameters and choices are fully presented and thatthe implications of the choices are explicitly discussed.

    LORETA (low resolution electromagnetic tomogra-phy) provides 3-dimensional functional imaging of brainelectrical activity based on multichannel surface EEG re-cording without a priori knowledge about the putativenumber of discernible source regions [84, 85]. It is impor-tant to notice that a reliable estimation of EEG sourcesdepends upon the usage of a sufficient number of elec-trodes (at least 19) which are equally distributed. The spa-tial resolution is limited by the distances between elec-

    trodes in the input matrix and by the assumption ofsmoothness. LORETA can be used to locate the mostprobable cortica l and subcortical sources for different fre-quency bands, and the time resolution of the EEG data canbe exploited for functional mapping of brain activities,whereby the solution space is restricted to cortical greymatter. Finally, LORETA images can also be computedrelative to normative voxel values, allowing sources to beevaluated as the deviation from age-expected norms.

    The empirical validity of LORETA has been estab-lished in a large number of studies [86] covering variousEEG research fields, such as psychiatry [87, 88], neurol-

    ogy [89, 90], neuropsychopharmacology [91] and sleepmedicine [92]. Further, the co-registration of LORETAwith other neuroimaging methods has shown good cor-respondence of effects [93, 94]. More recently, LORETAhas been used to evaluate interactive functional dynamicconnectivity in the brain, quantified by coherence andphase synchronization [95].

    Despite the limitations of inherently low resolutionand the need for a priori assumptions, source localisationtechniques represent important advances in the utility ofQEEG that are a useful addition to other available func-tional neuroimaging techniques, and EEG-based meth-

    ods provide direct information on neuronal activity withhigh temporal but low spatial resolution. In contrast,fMRI has high spatial resolution but low temporal resolu-tion that is confounded by the haemodynamic responsefunction. Magneto-encephalography combines high spa-tial and temporal resolution with a direct measure of ac-tivity but is less well established and not readily available.Also, it is sensitive to tangential and insensitive to radial

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    15/20

    IPEG Guidelines for Pharmaco-EEG Data Neuropsychobiology 2012;66:201220 215

    sources, whereas EEG is most sensitive to radial sources,so the techniques may provide different but complemen-tary information on brain activity [96].

    CordanceCordance is a QEEG analysis procedure that combines

    complementary information from both absolute and rel-ative power from the EEG spectrum, as well as informa-tion from neighbouring electrodes for each scalp elec-trode [97]. It has been reported to have a stronger correla-tion with cerebral perfusion as measured by positronemission tomography (PET) than either absolute or rela-tive power alone [98], potentially providing a physiologicframework for the interpretation of findings.

    Cordance has been used to assess neurophysiologicalchanges and treatment outcomes in major depression[99] and to predict the response of treatments in patientswith major depressive disorder [100102] or bipolar de-

    pression [103]. Across studies of patients with major de-pressive disorder treated with various antidepressantmedications, decreases in prefrontal cordance 1 weekafter the start of medication have consistently predictedresponse, with overall accuracy ranging from 72 to 88%.Examination of this measure in one randomized, double-blind, placebo-controlled trial has suggested it may be aspecific indicator of medication efficacy but not placeboefficacy [104].

    High-Frequency () EEG ActivityOver recent years, there has been considerably in-

    creased interest in EEG activities in the frequency do-main above 40 Hz. Prior to 2007, attempts to extract EEGactivity in the band used traditional analysis methods.However, in 2007, Whitham et al. [105] demonstratedthat a considerable proportion of thesignal extracted inthis way disappeared with temporary muscle paralysis.Subsequently, Yuval-Greenberg et al. [106] and Keren etal. [107] showed that most of the power in the widely re-ported induced broad-band peak at 200300 ms after avisual stimulus did not originate in the brain and was infact due to extra-ocular muscle activity during micro-saccades.

    Subsequently, methods have been developed to dealwith these tonic scalp and neck muscle and extra-ocularmuscle artefacts, hence enabling the true underlying EEGsignal to be extracted [107109]. These techniques requirerecording specifications that exceed those specifiedabove, including in particular: Increased sampling rate (25 kHz) Higher low-pass filter cut-off (5001,000 Hz)

    Improved amplitude resolution (0.1 V or below) Additional EOG electrodes to record fully the extra-

    ocular muscle activity Additional electrodes to record exogenous noise (e.g.

    power-line noise) for regression-based artefact reduc-tion

    Enhanced signal processing techniques are also needed,such as:

    Shorter epoch lengths (typically 512 or 256 ms) tomaintain the stationarity assumption of the FFT

    ICA- or regression-based methods for removal of ex-ogenous artefacts (e.g. power-line noise or pick-upfrom computer monitors) that would be removed bythe low-pass filter in traditional recordings

    ICA-, regression- or model-based methods for remov-al of tonic scalp and neck muscle and extra-ocularmuscle artefacts.This is an area of research in which techniques are de-

    veloping rapidly and there is insufficient evidence at pres-ent to determine the optimal recording and analysis tech-niques for high-frequency EEG. However, it is clear thatthe choice of method will quantitatively change the re-sults. Therefore, in studies including high-frequencyEEG, full details of the recording specification and anal-ysis techniques used, especially in relation to the artefactsdiscussed above, must be reported such that other labo-ratories can replicate, and potentially improve upon, theresults, so the field can continue to advance.

    In addition, the factor analysis used to define the fre-quency ranges presented in table 3 did not include cover-

    age of frequencies above 40 Hz. Hence, until a formal fac-tor analysis is undertaken to include the highfrequencyrange, it is important that studies clearly report the fre-quency bands that have been used. In the absence of anyother information, the following empirically chosen fre-quency ranges are suggested as a starting point for inves-tigators: 30 to !65 (1), 65 to !90 (2), and 90 to !135 Hz(3) [110, 111].

    EEG Biomarkers and Translational MedicineWithin the context of drug discovery, the definitions

    of a biomarker, clinical endpoint and surrogate endpoint

    have been formalised by the Biomarkers DefinitionsWorking Group [112]: A biomarker is a characteristicthat is objectively measured and evaluated as an indicatorof normal biological processes, pathogenic processes, orpharmacologic responses to a therapeutic intervention,whereas a surrogate endpoint is a biomarker that is in-tended to substitute for a clinical endpoint. Put differ-ently, a biomarker is an objectively measured index of

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    16/20

    Jobert et al.Neuropsychobiology 2012;66:201220216

    pharmacological response (or biological process) that isquantifiable, precise and reproducible. Biomarkers maybe used to diagnose or stage disease processes or predictclinical responses to treatments [113]. In the future, im-plementation of panels of relevant biomarkers may welltransform the process of development of new drugs [114].

    In the simplest case, EEG can be used as a PD bio-marker reflecting brain activity in healthy subjects with-out reference to a specific therapeutic indication. PK-PDmodelling can thus be undertaken in different species us-ing a similar outcome measure, hence enabling cross-species comparisons of the effects of a drug [115, 116].

    While EEG has a long history of evaluation in variousdiseases of the CNS, evidence is only recently accruing ofits putative validity as a useful and reliable biomarker forseveral therapeutic indications [14]. Recent reviews have,for instance, focussed on promising results indicating therelevance of QEEG as a reliable and sensitive biomarker

    in many areas, including: Prediction of antidepressant response [104] Early detection of mild cognitive impairment and

    Alzheimers disease [117, 118] Application in major neuropathophysiological disor-

    ders [119] Characterisation of sleep disturbances in depression

    [120] Assessment of vigilance in affective disorders [121] As a marker of genetic risk for attention deficit hyper-

    activity disorder [122] As a biomarker for localizing epileptogenic foci in the

    brain [123]In addition there is an increasing interest in the use of

    ERP biomarkers for characterisation of, for example, thecognitive disturbances in schizophrenia [124] or clinicaldepression [125].

    Another important aspect concerns the translatabilityof pharmaco-EEG signatures for the preclinical screen-ing and the development of CNS-active compounds. Thegoal of translational medicine applied to QEEG is tobring brain research from bench to bed and back. Thereare already several examples of successful contribution ofpreclinical EEG-based models in the discovery of psy-

    choactive drugs [14, 126, 127]. These EEG models in ani-mals have shown promise in improving the understand-ing of disease mechanisms and therapeutics (e.g. iden-tification of central effects at a functional level andcharacterisation of central effects depending on the psy-choactive class or therapeutic indication) or in studyingthe relationship between animal behaviour and EEG ac-tivity [128]. Recent papers have further shown the valid-

    ity of EEG as putative translatable biomarker in depres-sion [129] and Alzheimers disease [130].

    The major strength of using EEG in translationalmedicine is that most EEG parameters can be measuredin a similar way in various animal species and humansubjects, using broadly similar paradigms, recording

    techniques and signal processing algorithms: these simi-larities not only reflect face validity but also considerableconstruct validity. As similar QEEG methodology is ap-plicable in different species, it has the potential to bridgethe gap between preclinical and clinical research. How-ever, the lack of reliable translational procedures appli-cable to both patients and experimental animal models isan obstacle for the advancement of basic research, the de-termination of the translational validity of the technolo-gy and ultimately the development of new compounds[131] for three main reasons:(1) Lack of standardisation of animal EEG methodology

    (e.g. low lead density)(2) Lack of specific guidelines on animal QEEG and phar-

    maco-sleep EEG recording(3) Lack of valid animal models for psychiatric diseases.

    In this context, it becomes crucial that efforts are madein the standardisation of experimental conditions and inthe development of protocols facilitating the comparisonof data collected in both humans and animals. Such aconcerted harmonisation would also offer the potentialto quickly acquire large databases that could be sharedbetween research groups and hence enable a fuller studyand understanding of translatability, including:

    Equivalence of electrode specifications, positioningand montage across species including man

    Equivalence of preclinical and clinical paradigms,such as vigilance control for example

    Equivalence of spectral bands in different species(both numerically and in terms of species physiology)

    Differences between human scalp EEG recordings andpreclinical cortical recordings (including understand-ing differences between gyrencephalics and lissence-phalics)

    Equivalence and differences of underlying neurologi-cal processes and drug effects in different species and

    strains (from mice to primates)Specific guidelines for preclinical, animal pharmaco-

    EEG recording and analysis are in preparation for publi-cation by IPEG. Together with the present guidelines forstudies in man, these will address several of the above is-sues and hence should further contribute to optimisationof the translational value of QEEG.

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    17/20

    IPEG Guidelines for Pharmaco-EEG Data Neuropsychobiology 2012;66:201220 217

    Conclusion

    QEEG and related methods have the potential to offerreliable biomarkers and will, in view of the recent devel-opments in QEEG technology, play an increasingly im-portant role in all phases of drug development. The eval-

    uation and quantification of drug effects in pharmaco-EEG, sleep and EP/ERP studies provide a set of methodsto capture the therapeutic benefits and the potential ad-verse effects that a drug induces in diverse patient popu-lations. By combining various methods and their respec-

    tive strengths, it is reasonable to argue that they will pro-vide a more complete characterisation of the spectrum ofpharmacologic CNS responses [132].

    In this context, it is mandatory to enhance and stan-dardise methodology and facilitate comparability of dataacross laboratories both in academia and in industry. To

    this end, investigators using pharmaco-EEG are urged torefer to and comply with the guidelines presented herewhen designing and conducting studies, and to referencethe present paper when publishing study results.

    References

    1 Stille G, Herrmann WM: Guidelines forpharmaco-EEG studies in man; in Herr-mann WM (ed): Electroencephalography inDrug Research. Stuttgart, Fischer, 1982, pp

    1219.2 Dumermuth G, Ferber G, Herrmann WM,Hinrichs H, Knkel H: Recommendationsfor standardization of data acquisition andsignal analysis in pharmaco-electroenceph-alography. Neuropsychobiology 1987; 17:213218.

    3 Herrmann WM, Abt K, Coppola R, Eteve-non ET, Ferber G, Fink M, Gevins AS, Hin-richs H, Itil TM, John ER, Kubicki St, KnkelH, Kugler J, Lehmann D, Petsche H, Rappels-berger P, Rhmel J, Saito M, Saletu B, Scheu-ler W, Maurer K, Nuwer M: InternationalPharmaco-EEG Group (IPEG): recommen-dations for EEG and evoked potential map-ping. Neuropsychobiology 1989; 22: 170176.

    4 Pivik RT, Broughton RJ, Coppola R, DavidsonRJ, Fox N, Nuwer MR: Guidelines for the re-cording and quantitative analysis of electroen-cephalographic activity in research contexts.Psychophysiology 1993; 30: 547558.

    5 Versavel M, Leonard JP, Herrmann WM,Bttcher M, Dietrich B, Jobert M, Oldigs-Kerber J, Ott H, Reimann IW, Schaffler K,Schulz H, Wauschkuku HC, Gallhofer B,Lehmann D, Sannita W: Standard operatingprocedure (SOP) for the registration andcomputer-supported evaluation of pharma-co-EEG data. Working Team EEG in PhaseI of CIPS. Pharmacopsychiatry 1995; 28:245248.

    6 Duffy FH, Hughes JR, Miranda F, Bernad P,

    Cook P: Status of quantitative EEG (QEEG)in clinical practice. Clin Electroencephalogr1994; 25: 622.

    7 Nuwer MR: Assessment of digital EEG,quantitative EEG, and EEG brain mapping:report of the American Academy of Neurol-ogy and the American Clinical Neurophysi-ology Society. Neurology 1997; 49: 277292.

    8 Winterer G, Dorn H, Herrmann WM, Gall-hofer B, Bauer U, Hegerl U, Ihl R, Dierks T,Maurer K: The AMDP modules IIV: recom-mendations for a standardized acquisit ion of

    EEG data in psychiatry. Association forMethodology and Documentation in Psychi-atry. Neuropsychobiology 1997; 36: 100106.

    9 van Beijsterveldt CEM, van Baal GCM: Twinand family studies of the human electroen-cephalogram: a review and a meta-analysis.Biol Psychol 2002; 61: 111138.

    10 Smit DJ, Posthuma D, Boomsma DI, Geus EJ:Heritability of background EEG across thepower spectrum. Psychophysiology 2005; 42:691697.

    11 Smit CM, Wright MJ, Hansell NK, GeffenGM, Martin NG: Genetic variation of indi-

    vidual alpha frequency (IAF) and alpha pow-er in a large adolescent twin sample. Int J Psy-chophysiol 2006; 61: 235243.

    12 De Gennaro L, Marzano C, Fratello F, Mo-roni F, Pellicciari MC, Ferlazzo F, Costa S,Couyoumdjian A, Curcio G, Sforza E, Mala-fosse A, Finelli LA, Pasqualetti P, Ferrara M,Bertini M, Rossini PM: The electroencepha-lographic fingerprint of sleep is geneticallydetermined: a twin study. Ann Neurol 2008;64: 455460.

    13 Fink M: Remembering the lost neuroscienceof pharmaco-EEG. Acta Psychiatr Scand2010; 121: 161173.

    14 Leiser SC, Dunlop J, Bowlby MR, DevilbissDM: Aligning strategies for using EEG as asurrogate biomarker: a review of preclinicaland clinical research. Biochem Pharmacol2011; 81: 14081421.

    15 Brazier MAB: The effect of drugs on the elec-troencephalogram of man. Clin PharmacolTher 1964; 5: 102116.

    16 Fink M: Pharmaco-elec troencephalography:a note on its history. Neuropsychobiology1984; 12: 173178.

    17 Herrmann WM, Irrgang U: An absolutemust in clinico-pharmacological research:pharmaco-electroencephalography, its pos-sibilities and limitations. Pharmacopsychia-try 1983; 16: 134142.

    18 Itil T, Itil K: Quantitative EEG brain map-ping in psychotropic drug development,drug treatment selection, and monitoring.Am J Ther 1995; 2: 359367.

    19 Saletu B: The use of pharmaco-EEG in drugprofiling; in Hindmarch I, Stonier PD (eds):Human Psychopharmacology: Measuresand Methods. Chichester, Wiley, 1987, vol 1,pp 173200.

    20 Saletu B, Anderer P, Saletu-Zyhlarz GM, Ar-nold O, Pascual-Marqui RD: Classificationand evaluation of the pharmacodynamics ofpsychotropic drugs by single-lead pharma-co-EEG, EEG mapping and tomography(LORETA). Methods Find Exp Clin Pharma-col 2002; 24(suppl C):97120.

    21 Barbanoj MJ, Antonijoan RM, Riba J, ValleM, Romero S, Jan F: Quantifying drug-druginteractions in pharmaco-EEG. Clin EEGNeurosci 2006; 37: 108120.

    22 Knott VJ: Quantitative EEG methods andmeasures in human psychopharmacologicalresearch. Hum Psychopharmacol 2000; 15:479498.

    23 Barbanoj MJ, Riba J, Morte A, AntonijoanRM, Jan F: Basics of PK-PD using QEEG:acute/repetitive administration, interac-tions. Focus on anxiolytics with differentneurochemical mechanisms as examples.Methods Find Exp Clin Pharmacol 2002;24(suppl C):6783.

    24 Valle M, Barbanoj MJ, Jan F: Pharmaco-electroencephalography and pharmacoki-netic/pharmacodynamic modeling in basicresearch: focus on human pharmacology.Methods Find Exp Clin Pharmacol 2002;

    24(suppl D):129137.25 Barbanoj MJ, Valle M, Kulisevsky J, Prez V,Gambs P: Uses of pharmaco-EEG and phar-macokinetic-pharmacodynamic modelingin the clinical scenario. Methods Find ExpClin Pharmacol 2002; 24(suppl D):139144.

    26 Itil T: Quantitative pharmaco-electroen-cephalography; in Itil T (ed): PsychotropicDrugs and the Human EEG. Modern Prob-lems of Pharmacopsychiatry. Basel, Karger,1974, vol 8, pp 4375.

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    18/20

    Jobert et al.Neuropsychobiology 2012;66:201220218

    27 Galderisi S, Maj M, Mucci A, Bucci P, Ke-mali D: QEEG alpha 1 changes after a singledose of high-potency neuroleptics as a pre-dictor of short-term response to treatment inschizophrenic patients. Biol Psychiatry 1994;35: 367374.

    28 Mucci A, Volpe U, Merlotti E, Bucci P,Galderisi S: Pharmaco-EEG in psychiatry.

    Clin EEG Neurosci 2006; 37: 8198.29 ICH Guidelines. http://www.ich.org30 Wang T, Jacobson-Kram D, Pilaro AM, La-

    padula D, Jacobs A, Brown P, Lipscomb J,McGuinn WD: ICH guidelines: inception,revision, and implications for drug develop-ment. Toxicol Sci 2010; 118: 356367.

    31 Heather N: The importance of keeping regu-lar: accurate guidance to the public on low-risk drinking levels. Alcohol Alcohol 2009;44: 226228.

    32 Higdon JV, Frei B: Coffee and health: a re-view of recent human research. Crit RevFood Sci Nutr 2006; 46: 101123.

    33 Ulrich G, Frick K, Stieglitz RD, Mller-Oer-linghausen B: Interindividual variability of

    lithium-induced EEG changes in healthyvolunteers. Psychiatry Res 1987; 20: 117127.34 Bodenmann S, Rusterholz T, Drr R, Stoll C,

    Bachmann V, Geissler E, Jaggi-Schwarz K,Landolt HP: The funct ional Val158Met poly-morphism of COMT predicts interindividu-al differences in brainoscillations in youngmen. J Neurosci 2009; 29: 1085510862.

    35 Nuwer MR, Comi G, Emerson R, Fuglsang-Frederiksen A, Guerit J-M, Hinrichs H, Ike-da A, Luccas FJC, Rappelsburger P: IFCNstandards for digital recording of clinicalEEG. Electroencephalogr Clin Neurophysiol1998; 106: 259261.

    36 Jobert M: Recording EEG and EP data.Methods Find Exp Clin Pharmacol 2002;

    24(suppl C):716.37 Maus D, Epstein CM, Herman ST: DigitalEEG; in Schomer DL, Lopes da Silva FH (eds):Niedermeyers Electroencephalography: Ba-sic Principles, Clinical Applications, and Re-lated Fields, ed 6. Philadelphia, LippincottWilliams & Wilkins, 2011, pp 119141.

    38 Jasper HH: The ten-twenty electrode systemof the International Federation. Electroen-cephalogr Clin Neurophysiol 1958; 10: 371375.

    39 Chatrian GE, Lettich E, Nelson PL: Ten per-cent electrode system for topographic stud-ies of spontaneous and evoked EEG activi-ties. Am J EEG Technol 1985; 25: 8392.

    40 Nuwer MR: Recording electrode site nomen-

    clature. J Clin Neurophysiol 1987; 4: 121133.41 Oostenveld R, Praamstra P: The five percentelectrode system for high-resolution EEGand ERP measurements. Clin Neurophysiol2001; 112: 713719.

    42 Nunez PL, Srinivasan R: Recording strate-gies, reference issues, and dipole localiza-tion; in Nunez PL, Srinivasan R: ElectricFields of the Brain: The Neurophysics ofEEG, ed 2. New York, Oxford UniversityPress, 2006, pp 275312.

    43 Jobert M, Schulz H, Jhnig P: On the choice ofrecording duration in pharmaco-EEG studies.Neuropsychobiology 1995; 32: 106114.

    44 Olbrich S, Mulert C, Karch S, Trenner M,Leicht G, Pogarell O, Hegerl U: EEG-vigi-lance and BOLD effect during simultaneousEEG/fMRI measurement. NeuroImage2009; 45: 319332.

    45 Guenther T, Schnknecht P, Becker G, Ol-brich S, Sander C, Hesse S, Meyer PM, Lu-thardt J, Hegerl U, Sabri O: Impact of EEG-

    vigilance on brain glucose upta ke measuredwith [18F]FDG and PET in patients with de-pressive episode or mild cognitive impair-ment. Neuroimage 2011; 56: 93101.

    46 Croft RJ, Barry RJ: Removal of ocular artifactfrom the EEG: a review. Clin Neurophysiol2000; 30: 5 19.

    47 Joyce CA, Gorodnitsky IF, Kutas M: Auto-matic removal of eye movement and blinkartifacts from EEG data using blind compo-nent separation. Psychophysiology 2004; 41:313325.

    48 Schlgl A, Keinrath C, Zimmermann D,

    Scherer R, Leeb R, Pfurtscheller G: A fullyautomated correction method of EOG arti-facts in EEG recordings. Clin Neurophysiol2007; 118: 98104.

    49 Delorme A, Sejnowski T, Makeig S: En-hanced detection of arti facts in EEG data us-ing higher-order statistics and independentcomponent analysis. Neuroimage 2007; 34:14431449.

    50 Kemp B, Olivan J: European data formatplus (EDF+), an EDF alike standard formatfor the exchange of physiological data. ClinNeurophysiol 2003; 114: 17551761.

    51 Gasser T, Bcher P, Mcks J: Transforma-tions towards the normal distribution ofbroad band spectral parameters of the EEG.

    Electroencephalogr Clin Neurophysiol 1982;53: 119124.52 Koopman PAR, Wouters PAWM, Krijzer

    FNC: Mean power spectra from pharmaco-electrocorticographic studies: relative base-line correction and log transformation for aproper analysis of variance to assess drug ef-fects. Neuropsychobiology 1996; 33: 100105.

    53 Kiebel SJ, Tallon-Baudry C, Friston KJ: Para-metric analysis of oscillatory activity as mea-sured with EEG/MEG. Hum Brain Mapp2005; 26: 170177.

    54 Matejcek M: Vigilance and the EEG; in Herr-mann WM (ed): Electroencephalography inDrug Research. Stuttgart, Fischer, 1982, pp405508.

    55 Kubicki S, Herrmann WM, Fichte K, FreundG: Reflections on the topics: EEG frequencybands and regulation of vigilance. Pharma-kopsychiatr Neuropsychopharmakol 1979;12: 237245.

    56 Herrmann WM, Fichte K, Kubicki S: Defini-tion von EEG-Frequenzbnder aufgrundstrukturanalytischer Betrachtungen; in Ku-bicki S, Herrmann WM, Laudahn G (eds):Faktorenanalyse und Variablenbildung ausdem Elektroenzephalogramm. Stuttgart,Fischer, 1980.

    57 Herrmann WM: Electroencepha lography in

    Drug Research. Stuttgart, Fischer, 1982.58 Shackman AJ, McMenamin BW, Maxwell JS,

    Greischar LL, Davidson RJ: Identifying ro-bust and sensitive frequency bands for inter-rogating neural oscillations. Neuroimage2010; 51: 131933.

    59 Gasser T, Bcher P, Steinberg H: Test-retestreliability of spectral parameters of the EEG.Electroencephalogr Clin Neurophysiol 1985;60: 312319.

    60 Coppola R, Herrmann WM: Psychotropicdrug profiles: comparisons by topographicmaps of absolute power. Neuropsychobiolo-gy 1987; 18: 97104.

    61 Duff y FH, Bartels PH, Burchfiel JL: Signifi-cance probability mapping: an aid in the top-

    ographic analysis of brain electrical activity.Electroencephalogr Clin Neurophysiol 1981;51: 455462.

    62 Jhnig P, Jobert M: Methodological consid-erations for the evaluation of EEG mappingdata a practical example based on a place-bo/diazepam crossover trial. Neuropsycho-biology 1995; 31: 3146.

    63 Nuwer MR, Lehmann D, Lopes da Silva F,Matsuoka S, Sutherling W, Vibert J-F: IFCNguidelines for topographic and frequencyanalysis of EEGs and EPs. Report of an IFCNcommittee. International Federation ofClinical Neurophysiology. Electroencepha-logr Clin Neurophysiol 1994; 91: 15.

    64 Hjorth B: Source derivation simplifies topo-

    graphical EEG interpretation. Am J EEGTechnol 1980; 20: 121132.65 Michel CM, He B: EEG mapping and source

    imaging; in Schomer DL, Lopes da Silva FH(eds): Niedermeyers Electroencephalogra-phy: Basic Principles, Clinical Applications,and Related Fields, ed 6. Philadelphia, Lip-pincott Williams & Wilkins, 2011, pp 11791202.

    66 Soufflet L, Toussaint M, Luthringer R,Gresser J, Minot R, Macher JP: A statisticalevaluation of the main interpolation meth-ods applied to 3-dimensional EEG mapping.Electroencephalogr Clin Neurophysiol 1991;79: 393402.

    67 Herrmann WM, Abt K, Coppola R, Eteve-

    non ET, Ferber G, Fink M, Gevins AS, Hin-richs H, Itil TM, John ER, Kubicki S, KnkelH, Kugler J, Lehmann D, Petsche D, Rappels-berger P, Rhmel J, Saito M, Saletu B, Scheu-ler W: Recommendations for EEG andevoked potential mapping. Neuropsychobi-ology 1989; 22: 170176.

    68 Schoenknecht P, Olbrich S, Sander C, Spind-ler P, Hegerl U: Treatment of acute maniawith modafinil monotherapy. Biol Psychia-try 2010; 67:e55e57.

  • 8/12/2019 IPEG Guidelines for the Recording and Evaluation of Pharmaco-EEG Data in Man

    19/20

    IPEG Guidelines for Pharmaco-EEG Data Neuropsychobiology 2012;66:201220 219

    69 Hegerl U, Wilk K, Olbrich S, Schoenknecht P,Sander C: Hyperstable regulation of vigilancein patients with major depressive disorder.World J Biol Psychiatry 2012; 13: 436446.

    70 Olbrich S, Sander C, Jahn I, Eplinius F, ClausS, Mergl R, Schoenknecht P, Hegerl U: Un-stable EEG-vigilance in patients with can-cer-related fatigue (CRF) in comparison to

    healthy controls. World J Biol Psychiatry2012; 13: 146152.

    71 Ferber G, Abt K, Fichte K, Luthringer R:IPEG guideline on statistical design andanalysis for pharmacodynamic trials. Neu-ropsychobiology 1999; 39: 92100.

    72 Abt K: Significance testing of many variables problems and solutions. Neuropsychobiol-ogy 1983; 9: 4751.

    73 Abt K: Descriptive data analysis: a conceptbetween confirmatory and exploratory dataanalysis. Methods Inf Med 1987; 26: 7788.

    74 Abt K: Statistica l aspects of neurophysiolog-ic topography. J Cl in Neurophysiol 1990; 7:519534.

    75 Nichols TE, Holmes AP: Nonparametric

    permutation tests for functional neuroimag-ing: a primer with examples. Hum BrainMapp 2002; 15: 125.

    76 Maris E, Oostenveld R: Nonparametric sta-tistical testing of EEG- and MEG-data. JNeurosci Methods 2007; 164: 177190.

    77 John ER: Principles of neurometrics. Am JEEG Technol 1990: 30: 251266.

    78 Besserve M, Jerbi K, Laurent F, Baillet S,Martinerie J, Garnero L: Classificationmethods for ongoing EEG and MEG signals.Biol Res 2007; 40: 415437.

    79 Carozzo S, Fornaro S, Garbarino S, SaturnoM, Sannita WG: From neuroscience to ap-plication in neuropharmacology: a genera-tion of progress in electrophysiology. Clin

    EEG Neurosci 2006; 37: 121134.80 Hunter M, Smith RLL, Hyslop W, Rosso OA,Gerlach R, Rostas JAP, Williams DB, Hen-skens F: The Australian EEG database. ClinEEG Neurosci 2005; 36: 7681.

    81 John ER, Prichep LS, Friedman J, Easton P:Neurometrics: computer-assisted differen-tial diagnosis of brain dysfunctions. Science1988; 239: 162169.

    82 Prichep LS: Use of normative databases andstatistical methods in demonstrating clinicalutility of QEEG: importance and cautions.Clin EEG Neurosci 2005; 36: 8287.

    83 John ER, Prichep LS, Easton P: Normativedata banks and neurometrics: basic con-cepts, methods and results of norm con-

    struction; in Gevins AS, Remond A (eds):Methods of Analysis of Brain Electrical andMagnetic Signals. Handbook of Electroen-cephalography and Clinical Neurophysiolo-gy (Revised Series). Amsterdam, Elsevier,1987, pp 4 49495.

    84 Pascual-Marqui RD, Michel CM, LehmannD: Low resolution electromagnetic tomogra-phy: a new method for localizing electricalactivity in the brain. Int J Psychophysiol1994; 18: 4965.

    85 Pascual-Marqui RD: Standardized low-reso-lution brain electromagnetic tomography(sLORETA): technical details. Methods FindExp Clin Pharmacol 2002; 24(suppl D):512.

    86 Pascual-Marqui RD, Esslen M, Kochi K,Lehmann D: Functional imaging with lowresolution brain electromagnetic tomogra-phy (LORETA): review, new comparisons,

    and new validation. Jpn J Clin Neurophysiol2002; 30: 8194.

    87 Saletu B, Anderer P, Saletu-Zyhlarz GM, Pas-cual-Marqui RD: EEG mapping and low-res-olution brain electromagnetic tomography(LORETA) in diagnosis and therapy of psy-chiatric disorders: evidence for a key-lockprinciple. Clin EEG Neurosci 2005; 36: 108115.

    88 Saletu B, Anderer P, Saletu-Zyhlarz GM:EEG topography and tomography (LORE-TA) in diagnosis and pharmacotherapy ofdepression. Cl in EEG Neurosci 2010; 41: 203210.

    89 Saletu M, Anderer P, Saletu-Zyhlarz GM,Mandl M, Zeitlhofer J, Saletu B: Event-relat-

    ed potential low-resolution brain electro-magnetic tomography (ERP-LORETA) sug-gests decreased energetic resources for co