Personalized medicine in psychiatry is in need of biomarkers that resemble central nervous system function at the level of neuronal activity. Electroencephalography (EEG) during sleep or resting-state conditions and event-related potentials (ERPs) have not only been used to discriminate patients from healthy subjects, but also for the prediction of treatment outcome in various psychiatric diseases, yielding information about tailored therapy approaches for an individual. This review focuses on baseline EEG markers for two psychiatric conditions, namely major depressive disorder and attention deficit hyperactivity disorder. It covers potential biomarkers from EEG sleep research and vigilance regulation, paroxysmal EEG patterns and epileptiform discharges, quantitative EEG features within the EEG main frequency bands, connectivity markers and ERP components that might help to identify favourable treatment outcome. Further, the various markers are discussed in the context of their potential clinical value and as research domain criteria, before giving an outline for future studies that are needed to pave the way to an electrophysiological biomarker-based personalized medicine.

In some fields of medicine, individualized and personalized treatment has become state of the art. Especially in oncology, the assessment of individual biological properties of the patient and the cancer cells helped to make treatment more efficient, reduce side effects and improve secondary prevention strategies [1]. The paradigm shift from standard ‘one-size-fits-all' treatment plans according to descriptive markers such as stage and locus of the cancer to individual therapy algorithms based on e.g. genetic markers is thought to be of value also for neuropsychiatric disorders and raises new hopes for tailored therapies in psychiatry. However, a mental disorder is completely different from a well-observable and definable solid tumour: there is no clear organic correlate that is responsible for the symptoms; instead, multidimensional and possibly very heterogeneous alterations of brain function sum up to the clinical syndrome.

Although psychiatric disorders such as major depressive disorder (MDD) have an assumed high heritability of up to 37% [2], large-scale genome-wide association studies have thus far failed to link genetic variants with MDD [3]. This underpins the suggested polygenetic nature of psychiatric disorders [4] and implies the need for endophenotypes that are seen as an intermediate step between genotype and behaviour. Endophenotypes are more closely related to genotype than behaviour alone and may be a possible way to stratify a population for genome-wide association studies [5]. Although some promising findings using different sets of clinical and neuroimaging endophenotypes in major depression have been reported [6], a recent study on psychophysiological endophenotypes can be seen as a drawback to this approach since the authors were unable to replicate significant associations between endophenotypes and candidate genes [7].

Given that the link between endophenotypes and genetics might not be that strong or simple as suggested, an association between endophenotypes and disorder might still be present and could help to improve treatment and diagnostic decisions. In this context the term ‘biomarker' seems relevant, that is according to the National Institutes of Health ‘a characteristic that is objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes or pharmacologic responses to a therapeutic intervention' [8]. The value of a personalized medicine approach is not determined by the association of a marker with genetic variants but by the improvement it yields for the diagnostic process (by making it more objective) and, probably more importantly, by the increased effectiveness of the treatment (i.e. a more prognostic biomarker). As Thomas Insel, director of the National Institute of Mental Health, stated: ‘The task is to identify the biomarker that predicts response - whether the treatment is a medication or a psychosocial intervention.' The first step towards this ‘precision medicine' development was the introduction of the Research Domain Criteria project which is aimed to transform clinical syndrome-based diagnosis into an individualized framework of psychophysiology to support the diagnostic process of mental disorders [9] and - hopefully - improve treatment. Currently several large-scale studies are ongoing that should be able to shed more light onto this development, such as the international Study to Predict Optimized Treatment Response (iSPOT) in 2,016 patients with MDD and 672 children and adolescents with attention deficit hyperactivity disorder (ADHD; see also Williams et al. [10] for further protocol details) and the EMBARC study (Establishing Moderators and Biosignatures of Antidepressant Response for Clinical Care for Depression; https://clinicaltrials.gov/ct2/show/NCT01407094).

Sparked by the discovery that the mode of function of the human central nervous system is based on electric activity [11,12], the invention of electroencephalography (EEG) and its first description in humans [13] provided the possibility to analyse the brain at its core functional level. Taken as a tool for the assessment of biomarkers that, according to the definition, should be assessable objectively and provide information about physiological or pathological processes or responses to treatment interventions [14], EEG also fulfils the criteria of a cost-efficient, nowadays broadly available and already established tool in the diagnostic clinical practice. Further, EEG captures ongoing neuronal activity with a temporal resolution that surpasses any other neuroimaging modality such as functional magnetic resonance imaging or positron emission tomography. Also, the electroencephalogram is not a surrogate marker of neuronal activity (such as the blood deoxygenation level-dependent signal in functional magnetic resonance imaging or the glucose utilization in positron emission tomography; see Logothetis [15]) but is a direct reflection of neuronal activity (postsynaptic potentials) [16]. It is therefore highly plausible that a personalized medicine approach in psychiatry could gain from electrophysiological markers.

Although electrophysiological biomarkers have been studied throughout many psychiatric disorders, the current review is dedicated to only two of them: MDD and the ADHD. The following work describes the current state of the art of baseline EEG parameters by means of their diagnostic and predictive (prognostic) value. Treatment-emergent biomarkers that yield information about changes in the early course of treatment will not be subject of the review, and the interested reader is referred to Olbrich and Arns [17] or Arns and Olbrich [18] for more coverage of those.

EEG Sleep Research

EEG biomarkers in sleep are robust, and their advantage can be found in the link between clinical symptoms such as sleep initiation problems, early awakening or disrupted sleep in both MDD and ADHD and electrophysiologically assessable parameters. In MDD, the most consistently reported findings include a disturbed sleep architecture, comprising an increased rapid eye movement density [19,20], decreased rapid eye movement sleep latency [21,22] and altered slow-wave sleep in MDD [23,24].

While slow-wave power seems to have a discriminative value between MDD and healthy controls (HCs) [23,25], a predictive value for recurrence of depressive symptoms was found for decreased slow-wave sleep, decreased sleep efficiency and delayed sleep onset [26,27,28]. Also the slow-wave activity itself seems to be important for treatment prediction. Luthringer et al. [29] reported increased relative delta power in sleep EEG recordings in responders to antidepressant treatment, although others failed to replicate these findings [30,31]. Still, Nissen et al. [31] reported decreased slow-wave activity in responders, expressed in a high delta sleep ratio, a finding that again could not be replicated by Argyropoulos et al. [32]. Besides classical sleep EEG parameters, also a decreased coherence within the beta, delta and theta bands in sleep EEG predicted non-response in adolescents and the occurrence of depressive episodes [33].

In ADHD there is a clear lack of studies that examine EEG-derived sleep parameters, although other measures such as actigraphy and salivary melatonin measurements suggest a delayed sleep onset in a majority of children and adults with ADHD, also termed sleep onset insomnia [34,35,36] characterized by a delayed melatonin onset. This delayed sleep onset results in reduced sleep duration - and thus chronic sleep restriction - in ADHD, which becomes visible as the typical drowsiness patterns that can be observed in the EEG such as impaired vigilance (see EEG Vigilance below) or excess theta waves (see Frequency-Specific Biomarkers: Theta); for a review, see also Arns et al. [36] and Arns and Kenemans [37].

EEG Vigilance

Another possible EEG biomarker that has proven its value for differentiating between patients suffering from MDD and HCs is EEG vigilance regulation. Hegerl et al. [38] and Olbrich et al. [39] reported that MDD is associated with an increased EEG vigilance during rest with fewer and slower declines to lower vigilance stages during a 15-min resting condition. The appeal of this marker reflecting a high tone of CNS arousal can be found in the linkage between clinical symptoms and EEG parameters of wakefulness regulation. A hyperstable vigilance regulation in MDD is interpreted as an electrophysiological correlate of the often reported sleep problems. Increased vigilance might further explain the behavioural withdrawal of patients suffering from MDD to avoid a further increase in arousal [40].

The EEG vigilance framework further suggests that a fast decline of EEG vigilance during rest might result in increased sensation seeking and hyperactive behaviour to stabilize wakefulness regulation. Hegerl and Hensch [40] suppose that not only manic patients [41] reveal unstable EEG vigilance regulation patterns, but also patients suffering from ADHD [37,42]. In line with this, increased theta power as a marker of drowsiness has frequently been reported in patients with ADHD [43], and as described above the majority of patients with ADHD exhibit sleep onset insomnia [34,35,37], further supporting this notion. However, a recent meta-analysis also suggested increasing levels of theta power for healthy children and stable levels for ADHD children across the last 10 years [44], suggesting a possible gene × environment (sleep, circadian clock) interaction for this measure requiring further research.

Paroxysmal Patterns and Epileptic Discharges

Unlike in neurology, there are no distinct ‘grapho-elements' in EEG recordings that are pathognomonic for a psychiatric syndrome. However, already in 1939 was it demonstrated that during subclinical epileptiform activity patients had slower reaction times, while others did not respond at all [45], suggesting that paroxysmal activity in ‘non-epileptic patients' can have behavioural consequences.

The occurrence of paroxysmal EEG in affective disorders has not been investigated in much detail, but previous analyses suggest a prevalence of 3-5% in depression [46] to 20-40% in affective disorders, mostly mania [47]. The 3-5% in depression are comparable to the 1-6% prevalence of paroxysmal EEG in normal populations [47,48,49,50]. On the other hand, the occurrence of paroxysmal patterns in ADHD has been estimated to be between 12 and 15% [51,52,53] to approximately 30% [54], which is relatively high, compared to normal populations. A more recent study found epileptiform discharges in 25% of children with suspected ADHD [55].

The implications for treatment in psychiatric patients with paroxysmal patterns or epileptiform discharges - but without a history of seizures - remain unclear. It is still remarkable that several studies found that ADHD patients [56,57,58] do respond to anticonvulsant medication, e.g. to carbamazepine [59]. Furthermore, there is some evidence that antidepressant treatment augmentation with anti-epileptic drugs is effective in treatment resistant MDD [60,61], although data about the association of response and epileptiform discharges are lacking. As a further example, previous studies have demonstrated an association between paroxysmal EEG activity and panic attacks [for a review, see [62], [63]]. Patients with panic disorder and epileptiform EEG patterns have been found to clinically respond to anticonvulsants [64], thus suggesting that there could be a subgroup of psychiatric patients in whom paroxysmal/epileptiform EEG activity could be associated with their psychiatric complaints and for whom anticonvulsant treatment could be a choice for augmentation of treatment or as stand-alone treatment. However, this requires further controlled research.

Frequency-Specific Biomarkers: Alpha

One of the most prominent features of a resting-state EEG is the EEG alpha activity with heritability estimates of up to 79% [65]. Alpha activity in adults has a mean frequency around 10 Hz with a range between 7 and 13 Hz and has maximum amplitudes at parieto-occipital locations in the eyes-closed condition.

In MDD a consistent finding is an elevated absolute [66,67,68,69,70] or relative alpha power [71,72] at mainly parietal and frontal [68,73] or occipital sites [74]. The reason that some studies did not find alpha power differences between patients and HCs [75,76] or found decreased (relative) alpha activity in comparison to other patient groups [77] might be related to differences of recording periods, where shorter recording periods prevent the differences described above [38,39] in vigilance regulation to occur (e.g. 6 min in Knott and Lapierre [76] vs. 15 min in Hegerl et al. [38] and Olbrich et al. [39]).

In addition, there is some evidence that EEG alpha power can predict treatment outcome with low parieto-occipital [74,78,79] or lowered frontal alpha power [80] associated with non-response to antidepressants, although this could not be replicated in the recent multicentre iSPOT-D (for depression) study in 1,008 MDD patients [81]. However, for treatment with repetitive transcranial magnetic stimulation (rTMS), the opposite was reported [79,80], maybe related to higher levels of treatment resistance in these rTMS studies.

EEG alpha asymmetry has been investigated as a biomarker for MDD with a decreased alpha power at right frontal sites relative to the left side [82,83,84,85,86], although many studies have failed to replicate these findings [81,87,88,89,90,91]. Interestingly in ADHD, Keune et al. [92] found the opposite pattern of alpha asymmetry with increased right frontal alpha power.

Two studies by the same group investigated the prognostic value of alpha asymmetry in MDD and found conflicting results [74,93]; however, in the iSPOT-D study it was found that frontal alpha asymmetry (right frontal alpha dominance) was specifically related to response to the selective serotonin reuptake inhibitors escitalopram and sertraline, but not to the serotonin norepinephrine reuptake inhibitor venlafaxine in females only [81], underscoring the importance of large samples that allow testing for gender- and drug class-specific predictors of treatment outcome.

A slow background rhythm, also called a slow alpha peak frequency, has been consistently found a predictor for non-response to several treatments such as stimulant medication in ADHD [94], rTMS in depression [95,96] and the antidepressants pirlindol and amitriptyline [79]; for a review, see Arns [97].

Frequency-Specific Biomarkers: Theta

Several studies have reported elevated slow-wave activity in MDD [69,98,99,100,101], with the focus of this elevated theta activity localized to frontal areas and often to the anterior cingulate cortex (ACC) [68,101,102], though decreased ACC activity in MDD has also been reported [103] and some studies found no differences between MDD and controls [104,105].

High frontal theta activity has been associated with non-response to antidepressant treatments [96,106,107] while Cook et al. [108] found no differences. Seemingly contrary to this, Spronk et al. [109] reported that low theta activity at the frontal midline was associated with non-response. Note that several authors [96,106,107] reported on widespread frontal (not midline) theta activity, most likely a reflection of ‘drowsiness' theta power as discussed above (see EEG Vigilance) on vigilance [37], whereas Spronk and colleagues found the opposite pattern for frontal midline theta activity. This suggests these two types of theta activity could have different implications and different origins. In line with Spronk et al. [109], several studies have indeed shown low theta activity localized to the ACC, as estimated by source localization techniques, to respond worse to various antidepressant treatments [110,111,112,113]. These findings are in line with positron emission tomography and functional magnetic resonance imaging studies demonstrating low metabolic activity in the ACC is associated with worse treatment outcome; see also Pizzagalli [114] for an excellent review and meta-analysis on the rostral ACC (rACC) and treatment outcome. Contrary to this notion, several groups have reported high perfusion in the subcallosal cingulate (SCC; in earlier work this area was also referred to as rACC) [115] or rACC for non-responders [116,117], also reviewed in Arns et al. [101]. In line with this, recent results of the iSPOT-D study reported increased rACC and frontal theta activity to be associated with non-response, albeit with a small effect size [101]. Interestingly, these results tended to be driven mostly by treatment resistance, suggesting that future studies should also investigate the role of treatment resistance for the association of rACC/SCC perfusion and treatment outcome [101]. Conversely, deep-brain stimulation targeting the SCC in treatment- resistant MDD patients has been shown to result in clinical benefits [118], positing this area as a critical node in the depression network. However, the exact direction of these findings (high or low frontal midline theta activity) and exact localization (rACC vs. SCC) remains unclear from these lines of research, and future studies might shift the focus on investigating the connectivity of this specific area with other structures rather than focusing solely on EEG theta power given the above contradictory findings.

In ADHD, excess theta activity compared to controls is an often-reported finding, sometimes also expressed in the theta/beta ratio (TBR) [43], and several reports have termed the TBR a solid biomarker to identify ADHD. These findings resulted in this measure being FDA approved as a ‘diagnostic test' for ADHD [for a commentary, see [119]]. However, a recent meta-analysis could not confirm that this metric is a reliable ‘diagnostic test' for ADHD [41], due to an increased TBR across the last 10 years for controls, suggesting that this marker is a non-specific marker for drowsiness, and insufficient as a diagnostic biomarker for ADHD. Apart from the diagnostic use, this metric does hold potential as a prognostic biomarker being able to predict treatment outcome. A substantial proportion (26-38%) of ADHD subjects did have a high TBR and excess theta activity, and these subgroups have been found to be responders to stimulant medication [80,94,120] and neurofeedback [96,121], making this measure more likely a prognostic than diagnostic measure [119], albeit still requiring further replication.

Frequency-Specific Biomarkers: Beta

In MDD there is some evidence for increased beta EEG activity [99,122]. The predictive value for treatment outcome has not been investigated systematically so far.

In ADHD there is evidence for a subgroup of ADHD patients who are characterized by excess beta activity or beta spindles that make up 13-20% [123,124,125]. Several studies demonstrated that this subgroup does respond to stimulant medication [126,127,128]. A recent study further demonstrated that spindling excessive beta activity is a result of sleep maintenance problems and thus can indeed be considered a ‘subvigil' beta state and is specifically associated with impulse control problems, irrespective of diagnosis [129]. This would make the effectiveness of wakefulness-promoting drugs plausible in these patients.

EEG Connectivity Measures

First reports of altered connectivity in MDD in contrast to HCs stem from findings of altered coherence between EEG electrode sites [130,131]. More recent studies used a huge variety of connectivity measures like partial directed coherence, Granger causality, structural synchrony index and phase synchrony index. Some found decreased EEG connectivity in MDD [122,132,133,134] while others report of increased EEG connectivity in MDD, mainly in the alpha band [135,136,137,138]. More studies are needed to disentangle the complex relationship between the different connectivity measures and their physiological interpretation and to estimate the value for treatment prediction. In this context, one study [138] found an association between increased phase connectivity in the beta band between the subgenual prefrontal cortex and the right medial frontal cortex and treatment response. As suggested above in relation to theta activity, these approaches, when replicated, could further shed light onto the controversy between increased or decreased metabolism in the rACC/SCC.

Also in ADHD there is increasing evidence that EEG-based measures of connectivity could be used to differentiate between patients and HCs. Interestingly, most studies find increased measures of coherence especially within the beta and theta bands during the resting state [139,140,141]. Also graph theory network parameters seem to support an increased functional connectivity in ADHD [142]. Regarding a possible predictive value of connectivity measures, Dupuy et al. [143] describe an association between intrahemispheric coherence in the beta band and response to methylphenidate. These findings are promising and possibly pave the way for an improved differential diagnosis and consecutive treatment.

Event-Related Potentials

The event-related potential (ERP) is a waveform of averaged EEG activity, time-locked to a stimulus in a cognitive task. Several components of this ERP have been studied for their predictive value in treatment outcome.

In MDD research, the main focus has been on two measures, namely the P3 [144] and the loudness dependence auditory evoked potential (LDAEP), which is a derivative of the N1/P2 amplitude and its changes with increasing stimulus intensity [145]. So far, research involving the P3 has been ambiguous. Jaworska et al. [146] found that responders to antidepressants have larger P3 amplitudes than non-responders. A similar finding was reported by Bruder et al. [147] for the P3 amplitude at occipital sites. In contrast, responders to treatment with rTMS were found to have lower P3 amplitudes than non-responders, although this effect was limited to Pz and only marginally significant [96]. Regarding P3 latency, the results have been mixed as well, where some found no effect [146], while other studies found slower P3s in non-responders [148,149,150].

The LDAEP has proven to be a more robust predictor for antidepressant treatment response. A strong LDAEP, i.e. a steeper increase with stimulus intensity, is supposedly indicative of a low level of serotonergic activity [145,151] and is related to better outcome compared to a selective serotonin reuptake inhibitor [152], whereas the effect is reversed for responders to noradrenergic antidepressants [111,153,154]. A recent study, however, failed to replicate this relation between LDAEP and treatment outcome, even though a relation between treatment response and current source densities of the N1 LDAEP was obtained [155]. Related to this measure, Spronk et al. [109] found that a larger (more negative) N1 amplitude was related to a larger reduction of depressive symptoms after treatment with antidepressants.

In ADHD, ERP components have also been investigated as predictors for treatment response to stimulants. Sangal and colleagues found the topography of the P3, specifically the right frontocentral to parietal amplitude ratio, to be predictive of response to various stimulants, i.e. methylphenidate [156], atomoxetine [157] and pemoline [158]. They also reported a study in which poor responders to pemoline were treated with the antidepressant imipramine. Within this subgroup, poor responders to imipramine demonstrated slower P3 latencies [159]. Sunohara et al. [160 ]could not replicate ERP P3 and N2 latencies as baseline predictors for treatment outcome in ADHD children but found some treatment-emergent effects which are in agreement with Winsberg et al. [161].

Sleep EEG parameters have been found to be of discriminative and predictive value, especially in MDD. A widespread clinical use might be dampened due to the relatively large subject burden, e.g. one night at a sleep laboratory or even two to rule out first-night effects from findings [162]. In ADHD, there is a lack of studies that analyse EEG sleep parameters, although an association of the disorder with the sleep-wake cycle and especially circadian alterations is evident.

EEG vigilance measures seem to provide a less cost- and effort-intensive approach to assessing wakefulness regulation during rest in contrast to polysomnography. Another advantage of EEG vigilance-based markers can be found in its association with clinical features of MDD and ADHD as outlined above. A biomarker that has a direct link to the behavioural level is more likely to be accepted in clinical routine. Still, there is a clear lack of controlled studies that demonstrate the discriminative value of EEG vigilance parameters for response or non-response in MDD and in ADHD.

For patterns of paroxysmal or epileptiform activity, it remains unclear if treatment or augmentation with anti-epileptic drugs such as valproate, or some specific antidepressants, increase response rates in these MDD or ADHD subgroups. Some evidence exists that non-responders to first-line treatment may benefit. Studies that analyse the treatment of MDD or ADHD with anti-epileptic drugs should report on subgroups based on the occurrence of pathological but subclinical EEG patterns in the future.

Several quantitative EEG markers, especially within the alpha and theta range, revealed a discriminative value regarding treatment outcome. However, findings in this field are often contrary to what might be explainable by the variance of studied patient groups (treatment resistant or not) and at last not by the different treatment approaches that have been studied. Further, methodological and interpretation aspects need to be clarified as it is the case with the difference between diffuse frontal theta versus frontal-midline theta activity in MDD. Based on the existing findings, quantitative EEG measures of the alpha activity, alpha asymmetry and theta frequencies deserve to be in the main focus in future studies of treatment prediction.

EEG connectivity analysis has seen a revival within the past few years, and first findings seem to be promising with regard to its value for treatment prediction. However, the used measures differ broadly; there is almost no study that uses the same measures for the assessment of network interaction and connectivity. Therefore, the studies are hardly comparable. Further, many studies on EEG connectivity do not make an a priori hypothesis about alterations of connectivity between brain regions, resulting in a high number of tests that analyse every possible connectivity pattern, possibly resulting in type I errors. Identification of the most reliable and valid connectivity parameters and application on hypothesis-driven, predefined networks should be among the first goals of future research in this field.

Not only spontaneous EEG activity, but also ERPs hold value for the improvement of treatment. However, as it is the case in many resting-state EEG markers, there are promising markers that sometimes could not be replicated in small-scale studies. Therefore, larger study groups and controlled trials are needed to estimate the full potential of ERPs.

Since the aim of a personalized medicine approach is to improve treatment of the individual, studies are needed that analyse the predictive value of central nervous system arousal in patients for treatment outcome. Currently, data from the iSPOT and EMBARC trials in MDD are being analysed using this approach.

It should be noted that personalized medicine with a focus on treatment prediction is in need of addressing interindividual variance, which is in contrast to the search for biomarkers that reflect homogeneous diagnostic groups. Therefore, the mentioned differences in findings, sometimes even contrary to one another when looking at predictive markers for treatment response, might yield important information about different treatment options: it is possible that a marker predicts response to one treatment while it also could be found in non-responders to another treatment. The goal will be to disentangle these relationships with standardized and controlled trials, including a variety of treatment arms and by embracing heterogeneity.

The Research Domain Criteria provide a good framework to overcome subjective decisions in the treatment of psychiatric disorders and might help to bridge the gap to the advances made in medical treatment in other domains of medicine. In face of the myriads of available EEG-based parameters, it seems obvious that there will not be one single marker that fulfils all criteria to aid in the diagnosis and even predict treatment in different neuropsychiatric disorders. It will require combining a set of neurophysiological but also clinical and other biomarkers to fulfil the promise of a personalized medicine approach. Simulations on existing data sets and probably the usage of non-linear methods such as artificial networks could help in the analysis of large data matrices to extract meaningful combinations for treatment prediction [96]. It should be noted that the goal is not a final threshold or combination of biomarkers for prediction but a matrix of meaningful parameters that should be subject to further refinements. Furthermore, one fundamental problem to be faced is that of defining clinically meaningful treatment end points. Several different primary outcome measures have been used ranging from the Beck Depression Inventory-II (BDI-II) to the Hamilton Depression Inventory, and also using different criteria such as remission or response, whereas the agreement between these scales is far from perfect [163], thus making the ‘ground truth' of ‘clinical response' a moving target dependent on the instruments and definitions used.

In the first place it is now necessary to initialize biomarker-guided treatment decisions that lead to increased remission rates in comparison to treatment as usual. Furthermore, in MDD one faces the huge variety of treatment options, ranging from psychotherapy with different branches, over psychopharmacological interventions with completely different modes of action to brain stimulation methods such as TMS or electroconvulsive therapy but also sleep deprivation. In face of the high non-response rates, a framework is urgently needed including different biomarkers that allow for an evidence-based choice of the right treatment option at the right time for the right patients. Preferably markers should be taken into account that can be interpreted in the context of their physiological meaning since this will increase the acceptance of a marker by clinicians. Furthermore, as mentioned above concerning alpha asymmetry, large sample sizes are required to also address gender-, age- and drug class-specific predictors. Therefore, large multicentre studies for the identification of those markers and their thresholds to predict treatment outcome have to be carried out, as is the case with the iSPOT-D and EMBARC studies. The next step will then be to initiate prospective randomized controlled trials that compare the biomarker-based treatment versus treatment based on therapist and patient preferences as it is currently good clinical practice. This way, personalized medicine could help to apply the already existing treatment options in a hopefully more effective and efficient way and thereby decrease the individual burden of disease for patients.

1.
Weitzel JN, Blazer KR, Macdonald DJ, Culver JO, Offit K: Genetics, genomics, and cancer risk assessment: state of the art and future directions in the era of personalized medicine. CA Cancer J Clin 2011;61:327-359.
2.
Bienvenu OJ, Davydow DS, Kendler KS: Psychiatric ‘diseases' versus behavioral disorders and degree of genetic influence. Psychol Med 2011;41:33-40.
3.
Major Depressive Disorder Working Group of the Psychiatric GWAS Consortium, Ripke S, Wray NR, Lewis CM, Hamilton SP, Weissman MM, Breen G, Byrne EM, Blackwood DHR, Boomsma DI, Cichon S, Heath AC, Holsboer F, Lucae S, Madden PAF, Martin NG, McGuffin P, Muglia P, et al: A mega-analysis of genome-wide association studies for major depressive disorder. Mol Psychiatry 2013;18:497-511.
4.
Collins AL, Sullivan PF: Genome-wide association studies in psychiatry: what have we learned? Br J Psychiatry 2013;202:1-4.
5.
Gottesman II, Gould TD: The endophenotype concept in psychiatry: etymology and strategic intentions. Am J Psychiatry 2003;160:636-645.
6.
Glahn DC, Curran JE, Winkler AM, Carless MA, Kent JW, Charlesworth JC, Johnson MP, Göring HHH, Cole SA, Dyer TD, Moses EK, Olvera RL, Kochunov P, Duggirala R, Fox PT, Almasy L, Blangero J: High dimensional endophenotype ranking in the search for major depression risk genes. Biol Psychiatry 2012;71:6-14.
7.
Iacono WG, Vaidyanathan U, Vrieze SI, Malone SM: Knowns and unknowns for psychophysiological endophenotypes: integration and response to commentaries. Psychophysiology 2014;51:1339-1347.
8.
De Gruttola VG, Clax P, DeMets DL, Downing GJ, Ellenberg SS, Friedman L, Gail MH, Prentice R, Wittes J, Zeger SL: Considerations in the evaluation of surrogate endpoints in clinical trials. Summary of a National Institutes of Health workshop. Control Clin Trials 2001;22:485-502.
9.
Insel T, Cuthbert B, Garvey M, Heinssen R, Pine DS, Quinn K, Sanislow C, Wang P: Research Domain Criteria (RDoC): toward a new classification framework for research on mental disorders. Am J Psychiatry 2010;167:748-751.
10.
Williams LM, Rush AJ, Koslow SH, Wisniewski SR, Cooper NJ, Nemeroff CB, Schatzberg AF, Gordon E: International Study to Predict Optimized Treatment for Depression (iSPOT-D), a randomized clinical trial: rationale and protocol. Trials 2011;12:4.
11.
Caton R: Electrical currents of the brain.  J Nerv Ment Dis, 1875. http://journals.lww.com/jonmd/Fulltext/1875/10000/Electrical_Currents_of_the_Brain_.13.aspx.
12.
Ferrier D: Experiments on the brain of monkeys. No I. Royal Society of London, 1874. http://archive.org/details/philtrans02765954.
13.
Berger PDH: Über das Elektrenkephalogramm des Menschen. Arch Psychiatrie 1929;87:527-570.
14.
Atkinson AJ, Colburn WA, DeGruttola VG, DeMets DL, Downing GJ, Hoth DF, Oates JA, Peck CC, Schooley RT, Spilker BA, Woodcock J, Zeger SL: Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 2001;69:89-95.
15.
Logothetis NK: What we can do and what we cannot do with fMRI. Nature 2008;453:869-878.
16.
Buzsáki G, Anastassiou CA, Koch C: The origin of extracellular fields and currents - EEG, ECoG, LFP and spikes. Nat Rev Neurosci 2012;13:407-420.
17.
Olbrich S, Arns M: EEG biomarkers in major depressive disorder: discriminative power and prediction of treatment response. Int Rev Psychiatry 2013;25:604-618.
18.
Arns M, Olbrich S: Personalized medicine in ADHD and depression: use of pharmaco-EEG. Curr Top Behav Neurosci 2014;21:345-370.
19.
Goetz RR, Puig-Antich J, Dahl RE, Ryan ND, Asnis GM, Rabinovich H, Nelson B: EEG sleep of young adults with major depression: a controlled study. J Affect Disord 1991;22:91-100.
20.
Lauer CJ, Riemann D, Wiegand M, Berger M: From early to late adulthood. Changes in EEG sleep of depressed patients and healthy volunteers. Biol Psychiatry 1991;29:979-993.
21.
Reynolds CF 3rd, Kupfer DJ, Taska LS, Hoch CC, Spiker DG, Sewitch DE, Zimmer B, Marin RS, Nelson JP, Martin D: EEG sleep in elderly depressed, demented, and healthy subjects. Biol Psychiatry 1985;20:431-442.
22.
Rotenberg VS, Shami E, Barak Y, Indursky P, Kayumov L, Mark M: REM sleep latency and wakefulness in the first sleep cycle as markers of major depression: a controlled study vs schizophrenia and normal controls. Prog Neuropsychopharmacol Biol Psychiatry 2002;26:1211-1215.
23.
Hoffmann R, Hendrickse W, Rush AJ, Armitage R: Slow-wave activity during non-REM sleep in men with schizophrenia and major depressive disorders. Psychiatry Res 2000;95:215-225.
24.
Lopes MC, Quera-Salva M-A, Guilleminault C: Non-REM sleep instability in patients with major depressive disorder: subjective improvement and improvement of non-REM sleep instability with treatment (agomelatine). Sleep Med 2007;9:33-41.
25.
Armitage R, Emslie GJ, Hoffmann RF, Rintelmann J, Rush AJ: Delta sleep EEG in depressed adolescent females and healthy controls. J Affect Disord 2001;63:139-148.
26.
Emslie GJ, Armitage R, Weinberg WA, Rush AJ, Mayes TL, Hoffmann RF: Sleep polysomnography as a predictor of recurrence in children and adolescents with major depressive disorder. Int J Neuropsychopharmacol 2001;4:159-168.
27.
Hatzinger M, Hemmeter UM, Brand S, Ising M, Holsboer-Trachsler E: Electroencephalographic sleep profiles in treatment course and long-term outcome of major depression: association with DEX/CRH-test response. J Psychiatr Res 2004;38:453-465.
28.
Jindal RD, Thase ME, Fasiczka AL, Friedman ES, Buysse DJ, Frank E, Kupfer DJ: Electroencephalographic sleep profiles in single-episode and recurrent unipolar forms of major depression. II. Comparison during remission. Biol Psychiatry 2002;51:230-236.
29.
Luthringer R, Minot R, Toussaint M, Calvi-Gries F, Schaltenbrand N, Macher JP: All-night EEG spectral analysis as a tool for the prediction of clinical response to antidepressant treatment. Biol Psychiatry 1995;38:98-104.
30.
Buysse DJ, Hall M, Begley A, Cherry CR, Houck PR, Land S, Ombao H, Kupfer DJ, Frank E: Sleep and treatment response in depression: new findings using power spectral analysis. Psychiatry Res 2001;103:51-67.
31.
Nissen C, Feige B, König A, Voderholzer U, Berger M, Riemann D: Delta sleep ratio as a predictor of sleep deprivation response in major depression. J Psychiatr Res 2001;35:155-163.
32.
Argyropoulos SV, Hicks JA, Nash JR, Bell CJ, Rich AS, Nutt DJ, Wilson S: Redistribution of slow wave activity of sleep during pharmacological treatment of depression with paroxetine but not with nefazodone. J Sleep Res 2009;18:342-348.
33.
Morehouse RL, Kusumakar V, Kutcher SP, LeBlanc J, Armitage R: Temporal coherence in ultradian sleep EEG rhythms in a never-depressed, high-risk cohort of female adolescents. Biol Psychiatry 2002;51:446-456.
34.
Van Veen MM, Kooij JJS, Boonstra AM, Gordijn MCM, Van Someren EJW: Delayed circadian rhythm in adults with attention-deficit/hyperactivity disorder and chronic sleep-onset insomnia. Biol Psychiatry 2010;67:1091-1096.
35.
Van der Heijden KB, Smits MG, Van Someren EJW, Gunning WB: Idiopathic chronic sleep onset insomnia in attention-deficit/hyperactivity disorder: a circadian rhythm sleep disorder. Chronobiol Int 2005;22:559-570.
36.
Arns M, Feddema I, Kenemans JL: Differential effects of theta/beta and SMR neurofeedback in ADHD on sleep onset latency. Front Hum Neurosci 2014;8:1019.
37.
Arns M, Kenemans JL: Neurofeedback in ADHD and insomnia: vigilance stabilization through sleep spindles and circadian networks. Neurosci Biobehav Rev 2014;44:183-194.
38.
Hegerl U, Wilk K, Olbrich S, Schoenknecht P, Sander C: Hyperstable regulation of vigilance in patients with major depressive disorder. World J Biol Psychiatry 2012;13:436-446.
39.
Olbrich S, Sander C, Minkwitz J, Chittka T, Mergl R, Hegerl U, Himmerich H: EEG vigilance regulation patterns and their discriminative power to separate patients with major depression from healthy controls. Neuropsychobiology 2012;65:188-194.
40.
Hegerl U, Hensch T: The vigilance regulation model of affective disorders and ADHD. Neurosci Biobehav Rev 2014;44:45-57.
41.
Schoenknecht P, Olbrich S, Sander C, Spindler P, Hegerl U: Treatment of acute mania with modafinil monotherapy. Biol Psychiatry 2010;67:e55-e57.
42.
Sander C, Arns M, Olbrich S, Hegerl U: EEG-vigilance and response to stimulants in paediatric patients with attention deficit/hyperactivity disorder. Clin Neurophysiol 2010;121:1511-1518
43.
Boutros N, Fraenkel L, Feingold A: A four-step approach for developing diagnostic tests in psychiatry: EEG in ADHD as a test case. J Neuropsychiatry Clin Neurosci 2005;17:455-464.
44.
Arns M, Conners CK, Kraemer HC: A decade of EEG theta/beta ratio research in ADHD: a meta-analysis. J Atten Disord 2013;17:374-383.
45.
Kasteleijn-Nolst Trenité DG, Vermeiren R: The impact of subclinical epileptiform discharges on complex tasks and cognition: relevance for aircrew and air traffic controllers. Epilepsy Behav 2005;6:31-34.
46.
Arns MW, Kenemans Leon, Drinkenburg WHIM: University Utrecht personalized medicine in ADHD and depression : a quest for EEG treatment predictors. Utrecht University, 2011. http://dspace.library.uu.nl/handle/1874/215188.
47.
Shelley BP, Trimble MR, Boutros NN: Electroencephalographic cerebral dysrhythmic abnormalities in the trinity of nonepileptic general population, neuropsychiatric, and neurobehavioral disorders. J Neuropsychiatry Clin Neurosci 2008;20:7-22.
48.
Goodwin JE: The significance of alpha variants in the EEG, and their relationship to an epileptiform syndrome. Am J Psychiatry 1947;104:369-379.
49.
Lennox-Buchtal M, Buchtal F, Rosenfalck P: Correlation of electroencephalographic findings with crash rate of military jet pilots. Epilepsia 1960;1:366-372.
50.
Richter PL, Zimmerman EA, Raichle ME, Liske E: Electroencephalograms of 2,947 United States Air Force Academy cadets (1965-1969). Aerosp Med 1971;42:1011-1014.
51.
Satterfield JH, Cantwell DP, Saul RE, Lesser LI, Podosin RL: Response to stimulant drug treatment in hyperactive children: prediction from EEG and neurological findings. J Autism Child Schizophr 1973;3:36-48.
52.
Capute AJ, Niedermeyer EFL, Richardson F: The electroencephalogram in children with minimal cerebral dysfunction. Pediatrics 1968;41:1104.
53.
Hemmer SA, Pasternak JF, Zecker SG, Trommer BL: Stimulant therapy and seizure risk in children with ADHD. Pediatr Neurol 2001;24:99-102.
54.
Hughes JR, DeLeo AJ, Melyn MA: The electroencephalogram in attention deficit-hyperactivity disorder: emphasis on epileptiform discharges. Epilepsy Behav 2000;1:271-277.
55.
Millichap JJ, Stack CV, Millichap JG: Frequency of epileptiform discharges in the sleep-deprived electroencephalogram in children evaluated for attention-deficit disorders. J Child Neurol 2011;26:6-11.
56.
Itil TM, Rizzo AE: Behavior and quantitative EEG correlations during treatment of behavior-disturbed adolescents. Electroencephalogr Clin Neurophysiol 1967;23:81.
57.
Davids E, Kis B, Specka M, Gastpar M: A pilot clinical trial of oxcarbazepine in adults with attention-deficit hyperactivity disorder. Prog Neuropsychopharmacol Biol Psychiatry 2006;30:1033-1038.
58.
Silva RR, Munoz DM, Alpert M: Carbamazepine use in children and adolescents with features of attention-deficit hyperactivity disorder: a meta-analysis. J Am Acad Child Adolesc Psychiatry 1996;35:352-358.
59.
Wood JG, Crager JL, Delap CM, Heiskell KD: Beyond methylphenidate: nonstimulant medications for youth with ADHD. J Atten Disord 2007;11:341-350.
60.
Mowla A, Kardeh E: Topiramate augmentation in patients with resistant major depressive disorder: a double-blind placebo-controlled clinical trial. Prog Neuropsychopharmacol Biol Psychiatry 2011;35:970-973.
61.
Fang Y, Yuan C, Xu Y, Chen J, Wu Z, Cao L, Yi Z, Hong W, Wang Y, Jiang K, Cui X, Calabrese JR, Gao K; OPERATION Study Team: A pilot study of the efficacy and safety of paroxetine augmented with risperidone, valproate, buspirone, trazodone, or thyroid hormone in adult Chinese patients with treatment-resistant major depression. J Clin Psychopharmacol 2011;31:638-642.
62.
Boutros NN, Kirollos SB, Pogarell O, Gallinat J: Predictive value of isolated epileptiform discharges for a favorable therapeutic response to antiepileptic drugs in nonepileptic psychiatric patients. J Clin Neurophysiol 2014;31:21-30.
63.
Adamaszek M, Olbrich S, Gallinat J: The diagnostic value of clinical EEG in detecting abnormal synchronicity in panic disorder. Clin EEG Neurosci 2011;42:166-174.
64.
McNamara ME, Fogel BS: Anticonvulsant-responsive panic attacks with temporal lobe EEG abnormalities. J Neuropsychiatry Clin Neurosci 1990;2:193-196.
65.
Van Beijsterveldt CEM, van Baal GCM: Twin and family studies of the human electroencephalogram: a review and a meta-analysis. Biol Psychol 2002;61:111-138.
66.
Begić D, Popović-Knapić V, Grubišin J, Kosanović-Rajačić B, Filipčić I, Telarović I, Jakovljević M: Quantitative electroencephalography in schizophrenia and depression. Psychiatr Danub 2011;23:355-362.
67.
Grin-Yatsenko VA, Baas I, Ponomarev VA, Kropotov JD: Independent component approach to the analysis of EEG recordings at early stages of depressive disorders. Clin Neurophysiol 2010;121:281-289.
68.
Jaworska N, Blier P, Fusee W, Knott V: Alpha power, ALPHA asymmetry and anterior cingulate cortex activity in depressed males and females. J Psychiatr Res 2012;46:1483-1491.
69.
Roemer RA, Shagass C, Dubin W, Jaffe R, Siegal L: Quantitative EEG in elderly depressives. Brain Topogr 1992;4:285-290.
70.
Von Knorring L, et al: Intercorrelations between different computer-based measures of the EEG alpha amplitude and its variability over time and their validity in differentiating healthy volunteers from depressed patients. Adv Biol Psychiatry 1983;13:172-181.
71.
John ER, Prichep LS, Fridman J, Easton P: Neurometrics: computer-assisted differential diagnosis of brain dysfunctions. Science 1988;239:162-169.
72.
Prichep LS, John ER: qEEG profiles of psychiatric disorders. Brain Topogr 1992;4:249-257.
73.
Grin-Yatsenko VA, Baas I, Ponomarev VA, Kropotov JD: EEG power spectra at early stages of depressive disorders. J Clin Neurophysiol 2009;26:401-406.
74.
Bruder GE, Sedoruk JP, Stewart JW, McGrath PJ, Quitkin FM, Tenke CE: Electroencephalographic alpha measures predict therapeutic response to a selective serotonin reuptake inhibitor antidepressant: pre- and post-treatment findings. Biol Psychiatry 2008;63:1171-1177.
75.
Flor-Henry P, Koles ZJ, Howarth BC, Burton L: Neurophysiological studies of schizophrenia, mania and depression; in Gruzelier J, Flor-Henry P (eds): Hemisphere Asymmetries of Function in Psychopathology. New York, Elsevier/North Holland, 1979, pp 189-222.
76.
Knott VJ, Lapierre YD: Computerized EEG correlates of depression and antidepressant treatment. Prog Neuropsychopharmacol Biol Psychiatry 1987;11:213-221.
77.
Pozzi D, Golimstock A, Petracchi M, García H, Starkstein S: Quantified electroencephalographic changes in depressed patients with and without dementia. Biol Psychiatry 1995;38:677-683.
78.
Tenke CE, Kayser J, Manna CG, Fekri S, Kroppmann CJ, Schaller JD, Alschuler DM, Stewart JW, McGrath PJ, Bruder GE: Current source density measures of electroencephalographic alpha predict antidepressant treatment response. Biol Psychiatry 2011;70:388-394.
79.
Ulrich G, Renfordt E, Zeller G, Frick K: Interrelation between changes in the EEG and psychopathology under pharmacotherapy for endogenous depression. A contribution to the predictor question. Pharmacopsychiatry 1984;17:178-183.
80.
Suffin SC, Emory WH: Neurometric subgroups in attentional and affective disorders and their association with pharmacotherapeutic outcome. Clin Electroencephalogr 1995;26:76-83.
81.
Arns M, Bruder G, Hegerl U, Spooner C, Palmer D, Etkin A, Fallahpour K, Gatt J, Hieshberg L, Gordon E: EEG alpha asymmetry as a gender-specific predictor of outcome to acute treatment with different antidepressant medications in the randomized iSPOT-D study. Clin Neurophysiol, in press.
82.
Micoulaud-Franchi J-A, Richieri R, Cermolacce M, Loundou A, Lancon C, Vion-Dury J: Parieto-temporal alpha EEG band power at baseline as a predictor of antidepressant treatment response with repetitive transcranial magnetic stimulation: a preliminary study. J Affect Disord 2012;137:156-160.
83.
Chang JS, Yoo CS, Yi SH, Her JY, Choi HM, Ha TH, Park T, Ha K: An integrative assessment of the psychophysiologic alterations in young women with recurrent major depressive disorder. Psychosom Med 2012;74:495-500.
84.
Flor-Henry P: Lateralized temporal-limbic dysfunction and psychopathology. Ann NY Acad Sci 1976;280:777-795.
85.
Henriques JB, Davidson RJ: Left frontal hypoactivation in depression. J Abnorm Psychol 1991;100:535-545.
86.
Schaffer CE, Davidson RJ, Saron C: Frontal and parietal electroencephalogram asymmetry in depressed and nondepressed subjects. Biol Psychiatry 1983;18:753-762.
87.
Price GW, Lee JW, Garvey C, Gibson N: Appraisal of sessional EEG features as a correlate of clinical changes in an rTMS treatment of depression. Clin EEG Neurosci 2008;39:131-138.
88.
Carvalho A, Moraes H, Silveira H, Ribeiro P, Piedade RAM, Deslandes AC, Laks J, Versiani M: EEG frontal asymmetry in the depressed and remitted elderly: is it related to the trait or to the state of depression? J Affect Disord 2011;129:143-148.
89.
Gold C, Fachner J, Erkkilä J: Validity and reliability of electroencephalographic frontal alpha asymmetry and frontal midline theta as biomarkers for depression. Scand J Psychol 2013;54:118-126.
90.
Reid SA, Duke LM, Allen JJ: Resting frontal electroencephalographic asymmetry in depression: inconsistencies suggest the need to identify mediating factors. Psychophysiology 1998;35:389-404.
91.
Segrave RA, Cooper NR, Thomson RH, Croft RJ, Sheppard DM, Fitzgerald PB: Individualized alpha activity and frontal asymmetry in major depression. Clin EEG Neurosci 2011;42:45-52.
92.
Keune PM, Wiedemann E, Schneidt A, Schönenberg M: Frontal brain asymmetry in adult attention-deficit/hyperactivity disorder (ADHD): extending the motivational dysfunction hypothesis. Clin Neurophysiol 2015;126:711-720.
93.
Bruder GE, Stewart JW, Tenke CE, McGrath PJ, Leite P, Bhattacharya N, Quitkin FM: Electroencephalographic and perceptual asymmetry differences between responders and nonresponders to an SSRI antidepressant. Biol Psychiatry 2001;49:416-425.
94.
Arns M, Gunkelman J, Breteler M, Spronk D: EEG phenotypes predict treatment outcome to stimulants in children with ADHD. J Integr Neurosci 2008;7:421-438.
95.
Arns M, Spronk D, Fitzgerald PB: Potential differential effects of 9 Hz rTMS and 10 Hz rTMS in the treatment of depression. Brain Stimul 2010;3:124-126.
96.
Arns M, Drinkenburg WH, Fitzgerald PB, Kenemans JL: Neurophysiological predictors of non-response to rTMS in depression. Brain Stimul 2012;5:569-576.
97.
Arns M: EEG-based personalized medicine in ADHD: Individual alpha peak frequency as an endophenotype associated with nonresponse. J Neurother 2012;16:123-141.
98.
Kwon JS, Youn T, Jung HY: Right hemisphere abnormalities in major depression: quantitative electroencephalographic findings before and after treatment. J Affect Disord 1996;40:169-173.
99.
Lieber AL, Prichep LS: Diagnosis and subtyping of depressive disorders by quantitative electroencephalography. I. Discriminant analysis of selected variables in untreated depressives. Hillside J Clin Psychiatry 1988;10:71-83.
100.
Nyström C, Matousek M, Hällström T: Relationships between EEG and clinical characteristics in major depressive disorder. Acta Psychiatr Scand 1986;73:390-394.
101.
Arns M, Etkin A, Hegerl U, Williams LW, DeBattista C, Palmer D, Fitzgerald PB, Harris A, de Beuss R, Gordon E: Frontal and rostral anterior cingulate (rACC) theta EEG in depression: implications for treatment outcome? Eur Neuropsychopharmacol 2015, Epub ahead of print.
102.
Korb A, Cook I, Hunter A, Leuchter A: Brain electrical source differences between depressed subjects and healthy controls. Brain Topogr 2008;21:138-146.
103.
Mientus S, Gallinat J, Wuebben Y, Pascual-Marqui RD, Mulert C, Frick K, Dorn H, Herrmann WM, Winterer G: Cortical hypoactivation during resting EEG in schizophrenics but not in depressives and schizotypal subjects as revealed by low resolution electromagnetic tomography (LORETA). Psychiatry Res Neuroimag 2002;116:95-111.
104.
Lubar JF, Congedo M, Askew JH: Low-resolution electromagnetic tomography (LORETA) of cerebral activity in chronic depressive disorder. Int J Psychophysiol 2003;49:175-185.
105.
Pizzagalli DA, Nitschke JB, Oakes TR, Hendrick AM, Horras KA, Larson CL, Abercrombie HC, Schaefer SM, Koger JV, Benca RM, Pascual-Marqui RD, Davidson RJ: Brain electrical tomography in depression: the importance of symptom severity, anxiety, and melancholic features. Biol Psychiatry 2002;52:73-85.
106.
Iosifescu DV, Greenwald S, Devlin P, Mischoulon D, Denninger JW, Alpert JE, Fava M: Frontal EEG predictors of treatment outcome in major depressive disorder. Eur Neuropsychopharmacol 2009;19:772-777.
107.
Knott VJ, Telner JI, Lapierre YD, Browne M, Horn ER: Quantitative EEG in the prediction of antidepressant response to imipramine. J Affect Disord 1996;39:175-184.
108.
Cook IA, Leuchter AF, Witte E, Abrams M, Uijtdehaage SH, Stubbeman W, Rosenberg-Thompson S, Anderson-Hanley C, Dunkin JJ: Neurophysiologic predictors of treatment response to fluoxetine in major depression. Psychiatry Res 1999;85:263-273.
109.
Spronk D, Arns M, Barnett KJ, Cooper NJ, Gordon E: An investigation of EEG, genetic and cognitive markers of treatment response to antidepressant medication in patients with major depressive disorder: a pilot study. J Affect Disord 2011;128:41-48.
110.
Korb AS, Hunter AM, Cook IA, Leuchter AF: Rostral anterior cingulate cortex theta current density and response to antidepressants and placebo in major depression. Clin Neurophysiol 2009;120:1313-1319.
111.
Mulert C, Juckel G, Brunnmeier M, Karch S, Leicht G, Mergl R, Möller H-J, Hegerl U, Pogarell O: Prediction of treatment response in major depression: integration of concepts. J Affect Disord 2007;98:215-225.
112.
Narushima K, McCormick LM, Yamada T, Thatcher RW, Robinson RG: Subgenual cingulate theta activity predicts treatment response of repetitive transcranial magnetic stimulation in participants with vascular depression. J Neuropsychiatry Clin Neurosci 2010;22:75-84.
113.
Pizzagalli D, Pascual-Marqui RD, Nitschke JB, Oakes TR, Larson CL, Abercrombie HC, Schaefer SM, Koger JV, Benca RM, Davidson RJ: Anterior cingulate activity as a predictor of degree of treatment response in major depression: evidence from brain electrical tomography analysis. Am J Psychiatry 2001;158:405-415.
114.
Pizzagalli DA: Frontocingulate dysfunction in depression: toward biomarkers of treatment response. Neuropsychopharmacology 2011;36:183-206.
115.
Mayberg HS, Brannan SK, Mahurin RK, Jerabek PA, Brickman JS, Tekell JL, Silva JA, McGinnis S, Glass TG, Martin CC, Fox PT: Cingulate function in depression: a potential predictor of treatment response. Neuroreport 1997;8:1057-1061.
116.
McGrath CL, Kelley ME, Dunlop BW, Holtzheimer PE, Craighead WE, Mayberg HS: Pretreatment brain states identify likely nonresponse to standard treatments for depression. Biol Psychiatry 2014;76:527-535.
117.
Konarski JZ, Kennedy SH, Segal ZV, Lau MA, Bieling PJ, McIntyre RS, Mayberg HS: Predictors of nonresponse to cognitive behavioural therapy or venlafaxine using glucose metabolism in major depressive disorder. J Psychiatry Neurosci 2009;34:175-180.
118.
Mayberg HS, Lozano AM, Voon V, McNeely HE, Seminowicz D, Hamani C, Schwalb JM, Kennedy SH: Deep brain stimulation for treatment-resistant depression. Neuron 2005;45:651-660.
119.
Arns M, Gordon E: Quantitative EEG (QEEG) in psychiatry: diagnostic or prognostic use? Clin Neurophysiol 2014;125:1504-1506.
120.
Clarke AR, Barry RJ, McCarthy R, Selikowitz M: EEG differences between good and poor responders to methylphenidate and dexamphetamine in children with attention-deficit/hyperactivity disorder. Clin Neurophysiol 2002;113:194-205.
121.
Monastra VJ, Monastra DM, George S: The effects of stimulant therapy, EEG biofeedback, and parenting style on the primary symptoms of attention-deficit/hyperactivity disorder. Appl Psychophysiol Biofeedback 2002;27:231-249.
122.
Knott V, Mahoney C, Kennedy S, Evans K: EEG power, frequency, asymmetry and coherence in male depression. Psychiatry Res 2001;106:123-140.
123.
Chabot RJ, Serfontein G: Quantitative electroencephalographic profiles of children with attention deficit disorder. Biol Psychiatry 1996;40:951-963.
124.
Clarke AR, Barry RJ, McCarthy R, Selikowitz M: EEG-defined subtypes of children with attention-deficit/hyperactivity disorder. Clin Neurophysiol 2001;112:2098-2105.
125.
Clarke AR, Barry RJ, McCarthy R, Selikowitz M: EEG analysis in attention-deficit/hyperactivity disorder: a comparative study of two subtypes. Psychiatry Res 1998;81:19-29.
126.
Clarke AR, Barry RJ, McCarthy R, Selikowitz M, Clarke DC, Croft RJ: Effects of stimulant medications on children with attention-deficit/hyperactivity disorder and excessive beta activity in their EEG. Clin Neurophysiol 2003;114:1729-1737.
127.
Chabot RJ, Orgill AA, Crawford G, Harris MJ, Serfontein G: Behavioral and electrophysiologic predictors of treatment response to stimulants in children with attention disorders. J Child Neurol 1999;14:343-351.
128.
Hermens DF, Cooper NJ, Kohn M, Clarke S, Gordon E: Predicting stimulant medication response in ADHD: evidence from an integrated profile of neuropsychological, psychophysiological and clinical factors. J Integr Neurosci 2005;4:107-121.
129.
Arns M, Swatzyna RJ, Gunkelman J, Olbrich S: Sleep Maintenance, Spindling Excessive Beta and Impulse Control: An RDoC Arousal and Regulatory Systems Approach? Neuropsychiatr Electrophysiol 2015, in press.
130.
Lieber AL: Diagnosis and subtyping of depressive disorders by quantitative electroencephalography. II. Interhemispheric measures are abnormal in major depressives and frequency analysis may discriminate certain subtypes. Hillside J Clin Psychiatry 1988;10:84-97.
131.
O'Connor KP, Shaw JC, Ongley CO: The EEG and differential diagnosis in psychogeriatrics. Br J Psychiatry 1979;135:156-162.
132.
Lee T-W, Wu Y-T, Yu YW-Y, Chen M-C, Chen T-J: The implication of functional connectivity strength in predicting treatment response of major depressive disorder: a resting EEG study. Psychiatry Res 2011;194:372-377.
133.
Park C-A, Kwon R-J, Kim S, Jang H, Chae J-H, Kim T, Jeong J: Decreased phase synchronization of the EEG in patients with major depressive disorder; in Magjarevic R, Nagel JH (eds): World Congress on Medical Physics and Biomedical Engineering 2006. Berlin, Springer, 2007, pp 1095-1098.
134.
Sun Y, Li Y, Zhu Y, Chen X, Tong S: Electroencephalographic differences between depressed and control subjects: an aspect of interdependence analysis. Brain Res Bull 2008;76:559-564.
135.
Fingelkurts AA, Fingelkurts AA, Rytsälä H, Suominen K, Isometsä E, Kähkönen S: Impaired functional connectivity at EEG alpha and theta frequency bands in major depression. Hum Brain Mapp 2007;28:247-261.
136.
Jeong H-G, Ko Y-H, Han C, Kim Y-K, Joe S-H: Distinguishing quantitative electroencephalogram findings between adjustment disorder and major depressive disorder. Psychiatry Investig 2013;10:62-68.
137.
Leuchter AF, Cook IA, Hunter AM, Cai C, Horvath S: Resting-state quantitative electroencephalography reveals increased neurophysiologic connectivity in depression. PLoS One 2012;7:e32508.
138.
Olbrich S, Tränkner A, Chittka T, Hegerl U, Schönknecht P: Functional connectivity in major depression: increased phase synchronization between frontal cortical EEG-source estimates. Psychiatry Res 2014;222:91-99.
139.
González JJ, Méndez LD, Mañas S, Duque MR, Pereda E, De Vera L: Performance analysis of univariate and multivariate EEG measurements in the diagnosis of ADHD. Clin Neurophysiol 2013;124:1139-1150.
140.
Murias M, Swanson JM, Srinivasan R: Functional connectivity of frontal cortex in healthy and ADHD children reflected in EEG coherence. Cereb Cortex 2007;17:1788-1799.
141.
Barry RJ, Clarke AR: Resting state brain oscillations and symptom profiles in attention deficit/hyperactivity disorder. Suppl Clin Neurophysiol 2013;62:275-287.
142.
Barttfeld P, Petroni A, Báez S, Urquina H, Sigman M, Cetkovich M, Torralva T, Torrente F, Lischinsky A, Castellanos X, Manes F, Ibañez A: Functional connectivity and temporal variability of brain connections in adults with attention deficit/hyperactivity disorder and bipolar disorder. Neuropsychobiology 2014;69:65-75.
143.
Dupuy FE, Clarke AR, Barry RJ, McCarthy R, Selikowitz M: EEG coherence in children with attention-deficit/hyperactivity disorder: differences between good and poor responders to methylphenidate. Psychiatry Res 2010;180:114-119.
144.
Sutton S, Braren M, Zubin J, John ER: Evoked-potential correlates of stimulus uncertainty. Science 1965;150:1187-1188.
145.
Hegerl U, Juckel G: Intensity dependence of auditory evoked potentials as an indicator of central serotonergic neurotransmission: a new hypothesis. Biol Psychiatry 1993;33:173-187.
146.
Jaworska N, De Somma E, Blondeau C, Tessier P, Norris S, Fusee W, Smith D, Blier P, Knott V: Auditory P3 in antidepressant pharmacotherapy treatment responders, non-responders and controls. Eur Neuropsychopharmacol 2013;23:1561-1569.
147.
Bruder GE, Tenke CE, Stewart JW, Towey JP, Leite P, Voglmaier M, Quitkin FM: Brain event-related potentials to complex tones in depressed patients: relations to perceptual asymmetry and clinical features. Psychophysiology 1995;32:373-381.
148.
Işıntaş M, Ak M, Erdem M, Oz O, Ozgen F: Event-related potentials in major depressive disorder: the relationship between P300 and treatment response (in Turkish). Turk Psikiyatri Derg 2012;23:33-39.
149.
Kalayam B, Alexopoulos GS: Prefrontal dysfunction and treatment response in geriatric depression. Arch Gen Psychiatry 1999;56:713-718.
150.
Vandoolaeghe E, van Hunsel F, Nuyten D, Maes M: Auditory event related potentials in major depression: prolonged P300 latency and increased P200 amplitude. J Affect Disord 1998;48:105-113.
151.
Hegerl U, Gallinat J, Juckel G: Event-related potentials. Do they reflect central serotonergic neurotransmission and do they predict clinical response to serotonin agonists? J Affect Disord 2001;62:93-100.
152.
Gallinat J, Bottlender R, Juckel G, Munke-Puchner A, Stotz G, Kuss HJ, Mavrogiorgou P, Hegerl U: The loudness dependency of the auditory evoked N1/P2-component as a predictor of the acute SSRI response in depression. Psychopharmacology (Berl) 2000;148:404-411.
153.
Paige SR, Fitzpatrick DF, Kline JP, Balogh SE, Hendricks SE: Event-related potential amplitude/intensity slopes predict response to antidepressants. Neuropsychobiology 1994;30:197-201.
154.
Juckel G, Pogarell O, Augustin H, Mulert C, Müller-Siecheneder F, Frodl T, Mavrogiorgou P, Hegerl U: Differential prediction of first clinical response to serotonergic and noradrenergic antidepressants using the loudness dependence of auditory evoked potentials in patients with major depressive disorder. J Clin Psychiatry 2007;68:1206-1212.
155.
Jaworska N, Blondeau C, Tessier P, Norris S, Fusee W, Blier P, Knott V: Response prediction to antidepressants using scalp and source-localized loudness dependence of auditory evoked potential (LDAEP) slopes. Prog Neuropsychopharmacol Biol Psychiatry 2013;44:100-107.
156.
Sangal JM, Sangal RB: Attention-deficit/hyperactivity disorder: cognitive evoked potential (P300) topography predicts treatment response to methylphenidate. Clin Neurophysiol 2004;115:188-193.
157.
Sangal RB, Sangal JM: Attention-deficit/hyperactivity disorder: cognitive evoked potential (P300) amplitude predicts treatment response to atomoxetine. Clin Neurophysiol 2005;116:640-647.
158.
Sangal JM, Sangal RB, Persky B: Abnormal auditory P300 topography in attention deficit disorder predicts poor response to pemoline. Clin Electroencephalogr 1995;26:204-213.
159.
Sangal JM, Sangal RB, Persky B: Prolonged P300 latency in attention deficit hyperactivity disorder predicts poor response to imipramine. Clin Electroencephalogr 1996;27:191-201.
160.
Sunohara GA, Voros JG, Malone MA, Taylor MJ: Effects of methylphenidate in children with attention deficit hyperactivity disorder: a comparison of event-related potentials between medication responders and non-responders. Int J Psychophysiol 1997;27:9-14.
161.
Winsberg BG, Javitt DC, Silipo GS: Electrophysiological indices of information processing in methylphenidate responders. Biol Psychiatry 1997;42:434-445.
162.
Jobert M, Wilson FJ, Roth T, Ruigt GSF, Anderer P, Drinkenburg WHIM: Guidelines for the recording and evaluation of pharmaco-sleep studies in man: the International Pharmaco-EEG Society (IPEG). Neuropsychobiology 2013;67:127-167.
163.
Riedel M, Möller H-J, Obermeier M, Schennach-Wolff R, Bauer M, Adli M, Kronmüller K, Nickel T, Brieger P, Laux G, Bender W, Heuser I, Zeiler J, Gaebel W, Seemüller F: Response and remission criteria in major depression - a validation of current practice. J Psychiatr Res 2010;44:1063-1068.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.