Zum Hauptinhalt springen

Deep phenotyping towards precision psychiatry of first-episode depression - the Brain Drugs-Depression cohort.

Jensen, KHR ; Dam, VH ; et al.
In: BMC psychiatry, Jg. 23 (2023-03-09), Heft 1, S. 151
Online academicJournal

Deep phenotyping towards precision psychiatry of first-episode depression — the Brain Drugs-Depression cohort 

Background: Major Depressive Disorder (MDD) is a heterogenous brain disorder, with potentially multiple psychosocial and biological disease mechanisms. This is also a plausible explanation for why patients do not respond equally well to treatment with first- or second-line antidepressants, i.e., one-third to one-half of patients do not remit in response to first- or second-line treatment. To map MDD heterogeneity and markers of treatment response to enable a precision medicine approach, we will acquire several possible predictive markers across several domains, e.g., psychosocial, biochemical, and neuroimaging. Methods: All patients are examined before receiving a standardised treatment package for adults aged 18–65 with first-episode depression in six public outpatient clinics in the Capital Region of Denmark. From this population, we will recruit a cohort of 800 patients for whom we will acquire clinical, cognitive, psychometric, and biological data. A subgroup (subcohort I, n = 600) will additionally provide neuroimaging data, i.e., Magnetic Resonance Imaging, and Electroencephalogram, and a subgroup of patients from subcohort I unmedicated at inclusion (subcohort II, n = 60) will also undergo a brain Positron Emission Tomography with the [11C]-UCB-J tracer binding to the presynaptic glycoprotein-SV2A. Subcohort allocation is based on eligibility and willingness to participate. The treatment package typically lasts six months. Depression severity is assessed with the Quick Inventory of Depressive Symptomatology (QIDS) at baseline, and 6, 12 and 18 months after treatment initiation. The primary outcome is remission (QIDS ≤ 5) and clinical improvement (≥ 50% reduction in QIDS) after 6 months. Secondary endpoints include remission at 12 and 18 months and %-change in QIDS, 10-item Symptom Checklist, 5-item WHO Well-Being Index, and modified Disability Scale from baseline through follow-up. We also assess psychotherapy and medication side-effects. We will use machine learning to determine a combination of characteristics that best predict treatment outcomes and statistical models to investigate the association between individual measures and clinical outcomes. We will assess associations between patient characteristics, treatment choices, and clinical outcomes using path analysis, enabling us to estimate the effect of treatment choices and timing on the clinical outcome. Discussion: The BrainDrugs-Depression study is a real-world deep-phenotyping clinical cohort study of first-episode MDD patients. Trial Registration: Registered at clinicaltrials.gov November 15th, 2022 (NCT05616559).

Keywords: Major depressive disorder; Biomarker; Synaptic density; PET; MRI; SSRI; EEG; Cognition; Psychotherapy; Precision medicine

Supplementary Information The online version contains supplementary material available at https://doi.org/10.1186/s12888-023-04618-x.

Background

Depression is the leading cause of disability worldwide, with an estimated 322 million people currently suffering from a depressive episode [[1]]. We know that pharmacotherapy and psychotherapy, individually or combined, can efficaciously treat Major Depressive Disorder (MDD) [[2]].

Nevertheless, more than half of patients do not respond to the initial medication prescribed [[4]] and require sequential trials of different treatments that may not alleviate symptoms, resulting in treatment-resistant MDD [[5]]. After the first depressive episode, approximately half of patients will experience a relapse or re-emergence of depressive symptoms [[7]]. Current treatment guidelines emphasise depression severity as the primary or exclusive element on which to base treatment choice [[8]]. Despite numerous studies on definitions, frequency, and determinants of relapse and treatment-resistant MDD [[5], [9]], we continue to lack clinically relevant markers to guide treatment choice in first-episode MDD.

MDD is highly heterogeneous, with complex disease mechanisms and diverse biological and psychological causes [[10]], complicating drug discovery and optimal patient care. Deep phenotyping with a subsequent analysis to stratify patients' response to treatment is an essential tool to resolve this heterogeneity and move away from a 'one-size-fits-all' approach [[11]]. The prospect of this strategy is that patients instead can be stratified based on objective and replicable psychosocial, biochemical and/or neurobiological characteristics. Interventions would ideally be tailored to these individual profiles and thereby maximise clinical response [[12]]. Furthermore, precision medicine enables the optimisation of resource allocation within patient populations, e.g., by efficient allocation of relapse prevention and interventions to high-risk patient groups.

Previous studies have focused primarily on individual course predictors from just one or two domains, e.g., genetic [[13]], structural and functional neuroimaging [[14]–[18]], electroencephalographic biomarkers [[19]], blood markers [[21]], cognitive disturbances [[22]], and demographic or clinical data [[24]]. So far, no single marker has proven reliable enough to be implemented in clinical practice [[19], [26]]. A multimodal approach to depression is essential as the disorder's aetiology is likely multi-causal. That is, prediction models that combine multiple candidate biomarkers might have more predictive utility [[24]], but must still be generalizable and cross-validated across different cohorts [[27]]. Other issues include small sample sizes, using data from randomised control trials, lack of clinically and biologically relevant predictive markers, short follow-up time, and poorly validated and biased prediction models [[9], [26], [28]].

So far, most studies have focused on predictors of response to pharmacological antidepressant treatment without examining the effect of other commonly used treatment modalities such as psychotherapy. Factors such as safety, tolerability, and the effect of the patient's preferences (e.g., dislike of medication or psychotherapy) on overall treatment outcome are also underexplored [[26]]. Together with substantial methodological variance across studies, these limitations have hampered the ability to draw ecologically relevant and generalizable conclusions from meta-analyses [[26], [29]].

With this cohort-based study, we will generate a large longitudinal observational multi-modality clinical dataset that allows for a thorough analysis of which phenotypic components enable the best participant stratification for optimal treatment success. We will leverage the Danish healthcare system, which provides standardised treatment packages for all patients. Currently, group cognitive-behavioural therapy (CBT) constitutes the backbone of the treatment package with possibilities for initiation or changes in pharmacotherapy. The treatment package is nationally uniform and designed by Mental Health Services in the Capital Region. The treatment package was introduced in 2013 and revised in 2017 based on clinical and patient experience. However, while the treatment components in the package are research and evidence-based, the treatment effect in real life has not been examined.

The standardised treatment package system enables us to characterise newly diagnosed patients with depression and monitor their clinical response to different treatment modalities, combinations, and paths within the treatment package and deviations from it. Further, the individual Danish person identification number (CPR), combined with several national health and civil registers, will allow us to obtain additional information and follow the patients' clinical progress longitudinally [[31]].

Objectives

The primary objective of this study is to identify single or composite biomarkers that can reliably identify clinical profiles of MDD and predict their treatment outcomes.

To achieve this aim, we will establish a large, single-site cohort of adult patients diagnosed with first-episode MDD and referred for a treatment package for first-episode depression in secondary care, phenotyping patients before treatment initiation. All patients in the cohort (n = 800) will contribute with basic clinical, cognitive, psychometric, and biological data, i.e., genetics and blood biochemistry. A subcohort (subcohort I, n = 600) will provide Magnetic Resonance Imaging (MRI) and Electroencephalogram (EEG), and a second subcohort (subcohort II, n = 60) of subcohort I with patients unmedicated at inclusion will also undergo a Positron Emission Tomography (PET) brain scan with the presynaptic PET tracer [11C]-UCB-J. Clinical depression symptom severity is assessed with the Quick Inventory of Depressive Symptomatology (QIDS) at baseline (T0), and 6 (T1), 12 (T2) and 18 (T3) months after treatment initiation (Fig. 1). The treatment package typically lasts six months.

Graph: Fig. 1 Study design of the BrainDrugs-Depression prospective cohort. QoL Quality of life. Figure created by K.H.R.Jensen with BioRender.com

To examine disease trajectories, we will combine collected data with information from Danish national health and social registers, allowing us to further characterise and follow the patients before, during, and after treatment. Together, this might enable us to identify clinically relevant biomarkers and suggest treatment response algorithms, aiding treatment choices and improving patient care (Figure 1).

Furthermore, synaptic loss and deficits in functional connectivity are hypothesized to contribute to depressive symptoms, i.e., cognitive dysfunction, anhedonia, and anxiety and treatment effect. Therefore, we will examine the relationship between presynaptic density and cognitive dysfunction, depressive symptoms, and treatment effect in antidepressant naïve patients (subcohort II) and compare their cerebral presynaptic density to healthy controls (HC).

Hypotheses

The entire cohort

Primary hypotheses:

  • Clinical, cognitive, psychometric, genetic, and blood biomarker measures at inclusion can predict clinical remission (defined as QIDS ≤ 5) at the first follow-up.
  • Clinical, cognitive, psychometric, genetic, and blood biomarker measures at inclusion can predict clinical improvement (a ≥ 50% reduction in QIDS from pretreatment) at the first follow-up.

Secondary hypotheses:

  • Composite scores across a range of clinical, cognitive, psychometric, genetic, and blood biomarker measures at inclusion can cluster patients into MDD subgroups associated with treatment trajectories and outcomes.
  • Clinical, cognitive, psychometric, genetic, and blood biomarker measures at inclusion are associated with clinical outcome defined as a change in QIDS.
  • Path analysis of baseline patient characteristics and treatment tracks can uncover causal paths for clinical improvements, i.e., estimate the effect of treatment on clinical outcomes.
Subcohort I

Primary hypotheses:

  • MRI, fMRI, and EEG patterns at inclusion may be associated with depressive phenotypes.
  • Adding EEG, MRI, and fMRI measures at inclusion to the classifier model (defined in hypotheses 1.1 and 1.2) may significantly improve the prediction of clinical remission and improvement.

Secondary hypotheses:

  • Adding EEG, MRI, and fMRI measures at inclusion to the composite score (defined in hypothesis 1.3) may significantly improve the clustering of patients into MDD subgroups.
Subcohort II

Primary hypotheses:

  • Cerebral [11C]-UCB-J binding is lower in patients with MDD than in healthy controls.
  • Domain-specific cognitive function correlates positively with [11C]-UCB-J binding in associated cortical and subcortical areas.

Secondary hypotheses:

  • Depression severity, anxiety, and anhedonia correlate with [11C]-UCB-J binding in associated cortical and subcortical areas.
  • Addition of [11C]-UCB-J binding, EEG, and MRI measures at inclusion to the composite score (defined in hypotheses 2.1) can significantly improve the prediction of clinical improvement and remission beyond clinical, cognitive, psychometric, fluid biomarker, EEG, and MRI measures in antidepressant naïve patients.
Methods and design

Setting

The Capital Region of Denmark has a population of 1.6 million people. Patients in the Capital Region of Denmark are referred by their general practitioner (GP) or other treatment providers to a central diagnostic and referral centre within the mental health services that yearly assesses 20.000 referrals. About 4000 patients are further evaluated in person and diagnosed by the centre.

Five mental health centres in the region provide treatment packages for first-episode depression and will include participants in the study. The Mental Health Centre Amager and the Copenhagen centre consisting of two clinics located in the City of Copenhagen and treat approximately half of all patients, whereas Ballerup and Glostrup treat approximately a third of patients in the surrounding suburb (Sup. Figure 2). The Psychiatric Centre Northern Zealand treats approximately 16% of patients and is located north of Copenhagen, in a region of intermediate urbanisation with individual municipalities classified as rural.

Study population

We aim to establish a cohort of 800 patients referred to the Danish treatment packages for unipolar first-episode, non-psychotic depression during 2021–2025. We recruit patients from all six clinics in the region. Each clinic receives approximately 100–250 treatment referrals yearly, and approximately 1100 patients are referred yearly. Approximately 80% of referrals are sent directly to the clinics. Patients are recruited during evaluation at the central diagnostic and referral centre or the first consultation in the clinics. Approximately 88% of referrals result in treatment package initiation.

During 2019–2020, 37% of patients were on an antidepressant (usually the selective serotonin reuptake inhibitor (SSRI) Sertraline from their GP) when starting the treatment package, and 54% of patients ended the treatment package on an antidepressant medication. 13% of patients were transferred to a treatment package for a different primary diagnosis group, e.g., generalised, social anxiety, post-traumatic stress disorder, emotionally unstable personality, avoidant personality disorder, eating disorder or obsessive–compulsive disorder. 20% dropped out of treatment. 5% of patients were hospitalised during their treatment package; hospitalization does not preclude the continuation of the treatment package.

The treatment package is a program with manualised psychotherapy in groups of eight patients as the core treatment module together with psychoeducation for the patient and relative (Sup. Table 1). In brief, a treatment package consists of 15–18 h: 2–3 h of initial workup followed by 6 h of individual therapy or 12 sessions of 2 h group therapy (8 patients per group); 1–2 h of engagement and psychoeducation of relatives; 1–5 h of medication clinic; and 2 h of relapse prevention. The program is designed around group-based CBT, but clinics also offer alternatives to CBT, e.g., psychodynamic and schema therapy, and groups for specific demographics, e.g., men or adolescents, and individual therapy. Medication is available as needed.

The research and assessment at baseline for recruited participants is conducted at the Neurobiology Research Unit (NRU) at the Copenhagen University Hospital Rigshospitalet and followed by clinicians from the Mental Health Centre Copenhagen who are not involved in the patient's treatment.

Inclusion and exclusion criteria for patients

Patients between 18 and 65 years of age referred to a treatment package for single-episode depression will be recruited (Table 1) with minimal exclusion criteria to recruit representative adult outpatients who would typically receive treatment in routine practice (Table 1), of which the majority are women (71%) and aged 18–35 (68%) (S. Figure 1). Patients over 65 (approximately 0.7% of the target population) are excluded because of potential age-related cognitive decline, concomitant medical conditions, or medications that could interact with assessments or treatment (S. Figure 1). Allocation into the subcohorts is based on eligibility, e.g., MRI compatibility, scheduling, and patient willingness to participate.

Table 1 Inclusion and exclusion criteria for patients

Patient inclusion criteria:

• Fulfilment of ICD-10 diagnostic criteria for a primary depressive episode (i.e., not secondary to known organic or other psychiatric disorder)

• Referral to a treatment package for single-episode depression

• Age between 18 and 65 years

Exclusion criteria:

• Psychosis or psychotic symptoms

• History of severe head trauma involving hospitalization or unconsciousness for more than 5 min

• Known, substantial structural brain abnormalities

• Insufficient Danish language skills to complete questionnaires and cognitive testing

Additional exclusion criteria for subcohort I:

• Severe somatic disease

• Contraindications for MRI (e.g., metal implants, claustrophobia, or back problems)

Additional exclusion criteria for subcohort I:

• Use of psychotropic drugs

• Exposure to radioactivity > 10 mSv within the last year

• Pregnancy or breastfeeding

The primary depressive episode, consistent with the International Statistical Classification of Diseases and Related Health Problems version 10 (ICD-10) criteria for MDD without psychotic features (F32.1, F32.2, F32.8 and F32.9), is confirmed by a specialist in psychiatry at the central diagnostic and referral centre.

Inclusion and exclusion criteria for healthy controls

Data from HCs for comparisons to patients with MDD are available on-site from recent and concurrent projects, stored in the Cimbi database described in [[32]] including the BrainDrugs-Epilepsy study [[33]]. Apart from psychiatry-related issues (e.g., no current or history of mental illness or unstable somatic condition), the HC meet the same inclusion and exclusion criteria as required for patients.

Data collection

The patients will undergo a multi-modal investigative program at inclusion and will be followed up after treatment and 12 and 18 months after treatment initiation with questionnaires assessing clinical status (Figs. 1 and 2). Apart from follow-up measures and information extracted from patient files and Danish health and social registries, all data collection will occur before the patient starts treatment.

Graph: Fig. 2 Flow diagram (STROBE) of the BrainDrugs-D cohort study. CVD: (Center for Visitation og Diagnostik) the central referral center within the mental health services in the capital region of Denmark, QIDS-SR: Quick Inventory of Depressive Symptomatology – Self Report, BSI-18: 10 item Brief Symptom Inventory, SCL-10: 10 item Symptom Checklist, mSDS: Modified Sheehan Disability Score, WHO-5: World Health Organisation—Five Well-Being Index (WHO-5), NEQ: Negative Effects Questionnaire, PRISE: Patient-Related Inventory of Side Effects

Baseline

Questionnaires

Questionnaires will be completed through a secure, web-based survey system hosted by the research centre so that participants can complete questionnaires electronically, either at home or during their visit to the research centre. Online questionnaires are sent via a national secure mail platform used by citizens in regular correspondence with public institutions and the health care system.

Measures include several salient domains in the clinical characterisation of the patient, among others, assessments of demographics (e.g., ethnicity, education, and marital status); medical and psychiatric history; depressive symptoms and impact of depression behaviour and day-to-day life; treatment preferences and expectations, life experiences; and a broad range of state and trait psychometrics. Some questionnaires will only be given to patients in subcohorts I-II (Table 2).

Table 2 Questionaries Additional questionnaires for the subcohort I-II only are in bold

Symptom profile and Severity

Cognitive style

Upbringing and life history

Functioning and quality of life

Inventory of Depressive Symptomatology – self-report (IDS-SR) [34]

Mentalisation Questionnaire (MZQ) [35]

Online Stimulant and Family History Assessment Module (OS-FHAM) [11]

Cognitive Complaints in Bipolar Disorder Rating Assessment (COBRA) [36]

Dimension of Anger Reactions (DAR-5) [37]

Ruminative Response Scale (RRS) [38]

Child abuse and trauma scale (CATS) [39]

Modified Sheehan Disability Score (mSDS)

Generalised Anxiety Disorder 7-item (GAD-7) [40]

Perth Alexithymia Questionnaire (PAQ) [41]

Parental Bonding Instrument (PBI) [42]

WHO 5 wellbeing index (WHO-5)

Cohen's Perceived Stress Scale (PSS) [43]

Mindful Attention Awareness Scale (MAAS) [44]

Stressful Life Events (SLE) [45]

Changes in Sexual Functioning Questionnaire (CSFQ) [46]

Brief Symptom Inventory (BSI) [47]

Short form of Metacognitions Questionnaire (MCQ-30) [48]

Questions from the Copenhagen Aging and Midlife Biobank (CAMB) [49]

Symptom checklist (SCL-10) [50]

Coping Self-Efficacy Scale (CSES) [51]

Revised Sociosexual Orientation Inventory (SOI-R) [52]

Snaith-Hamilton Pleasure Scale (SHAPS) [53]

Pittsburgh Sleep Quality Index (PSQI) [54]

Medical records and registry data

Detailed medical information about previous illness, medication usage, hereditary dispositions, drug, tobacco, and alcohol intake will be acquired for all participants through interviews, self-report questionnaires, electronic medical records (EMR), and registry data.

Data extracted from the EMR will include treatment codes from the MDD treatment package, dates for treatment package start and completion, psychiatric comorbidities; and standard clinical blood work (e.g., HBA1c, TSH, CRP, and cholesterol). In addition, hormonal contraceptive and psychotropic medication prescription and usage (from 1995 onward) will be extracted from the Danish National Prescription Registry [[55]]. This information includes prescribed medication and dosage and when the patient redeems a prescription. We will retrieve information on lifetime comorbidity from The Danish National Patient Registry (DNPR) [[57]]. From the Medical Birth Registry, we will obtain data on maternal and maternal perinatal health [[58]]. We will also collect information on alcohol and drug abuse treatment from the National Registry of Alcohol Treatment and Registry of Drug Abusers Undergoing Treatment. From the social registers in Statistics Denmark, we add data on marital status, occupational history, ethnicity, and educational level [[59]].

Cognitive testing

All patients are assessed with a ~ 1-h neuropsychological test battery, including 'cold' (emotion-independent) cognitive tasks indexing reaction time; psychomotor speed; verbal learning and memory; working memory; and executive functions, as well as 'hot' (emotion-dependent) cognitive tasks from the Danish version of the EMOTICOM test-battery indexing emotion recognition; emotion detection; and moral emotions in social situations [[60]].

Patients in subcohorts I-II will complete an additional ~ 1 h of testing with tasks assessing mental flexibility, verbal fluency, and visuospatial learning and memory (see Additional questionnaires for the subcohort I-II only are in bold.

Table 3 for a complete overview of all cognitive tasks). In addition, patients' subjective experiences of cognitive disturbances will be assessed by the Cognitive Complaints in Bipolar Disorder Rating Assessment (COBRA) questionnaire [[36]].

Table 3 Cognitive testing before treatment

Cognitive Test

Cognitive Domaine

Whole Cohort

Simple Reaction Time task (SRT)

Reaction time

Trail Making Test A & B

Psychomotor speed/executive function

Symbol Digit Modality Task (SDMT)

Psychomotor speed/working memory

Letter-Number Sequence (LNS)

Working memory

D-KEFS Color-Word Interference Test (Stroop)

Executive function

Rey Auditory Verbal Learning Test (RAVLT)

Learning/memory

EMOTICOM Emotional Recognition Task (ERT)

Emotion recognition accuracy

EMOTICOM Emotional Intensity Morphing Task (IMT)

Emotion perceptual detection threshold

EMOITCOM Moral Emotions Task (MET)

Social cognition: guilt and shame

Additional testing in the subcohorts

D-KEFS Verbal Fluency

Executive function

Rey Complex Figure Test (RCFT)

Visuo-spatial learning/memory

Probabilistic Reversal Learning task

Learning within a feedback context

Screen for Cognitive Impairments in Psychiatry—Depression (SCIP-D)

Memory, working memory, vocabulary, psychomotor speed

Patients in subcohorts I-II will complete an additional ~ 1 h of testing with tasks assessing mental flexibility, verbal fluency, and visuospatial learning and memory (see Additional questionnaires for the subcohort I-II only are in bold.

Blood biochemistry, genetics, and gene expression

Venous blood samples will be collected for serum, plasma, DNA, and RNA extraction. Identifying biomarkers relevant to the course of depression is an area of research that is evolving rapidly. Thus, based on an ongoing critical literature review, the search for and analysis of specific biomarkers may change during the study period. Currently, the blood biomarkers include inflammation parameters (e.g., high sensitivity CRP) [[61]] and neurotrophic factors (e.g., BDNF and S100B) [[63]].

DNA from blood samples will be used for microarray-based genotyping of MDD candidate genes, genes of relevance for MDD (e.g., rs41271330, 5-HTTLPR, COMT, and BDNFval66met), drug metabolism (e.g., CYP2D6, CYP2C19, UGT1A1, ABCB1, ABCC1) and to compute polygenic risk scores in all participants after genome-wide genotyping in the future. DNA will also be used for epigenetic analysis, and circular extrachromosomal DNA, a form of decomposed free DNA [[65]], will be extracted and characterised. RNA will be extracted for gene transcription profiles using microarray or TAG-based methods (mRNA and microRNA).

DNA from blood samples will be used for microarray-based genotyping of MDD candidate genes, genes of relevance for MDD (e.g., rs41271330, 5-HTTLPR, COMT, and BDNFval66met), drug metabolism (e.g., CYP2D6, CYP2C19, UGT1A1, ABCB1, ABCC1) and to compute polygenic risk scores in all participants after genome-wide genotyping in the future. DNA will also be used for epigenetic analysis, and circular extrachromosomal DNA, a form of decomposed free DNA [[65]], will be extracted and characterised. RNA will be extracted for gene transcription profiles using microarray or TAG-based methods (mRNA and microRNA).

Gene analyses will be based on a priori models of genetic variations known to modulate pharmacotherapy and psychotherapy responses. The results will be used to calculate a polygenic risk score for diagnosis and treatment response and meta-analyses with established polygenic risk scores for MDD and those currently developed for anxiety and anxiety disorders, including treatment response [[66]].

Electroencephalogram

In subcohort I, we will record resting state EEG and event-related potentials (ERPs) with simultaneous two-lead electrocardiography (ECG) to measure autonomic nervous system activation. EEG will be recorded using a 256-channel HydroCel Sensor Net system (MagstimEGI, USA) at 1000 Hz, where the vertex electrode serves as the reference. Impedances across all electrodes will be kept below 50 kΩ. ECG will be acquired at 1000 Hz using a Physio 16 device (MagstimEGI, USA). EEG/ERP recording: resting EEG (6 min eyes closed and eyes open), two-tone auditory oddball and the LDAEP tasks.

MRI

Participants in subcohort I will undergo MRI using a Siemens 3-Tesla Magnetom Prisma scanner. High-resolution structural T1-, T2-, and diffusion-weighted MR images will be acquired as well as ultra-fast functional magnetic resonance encephalography (MREG) asses cardiovascular brain pulsations [[67]]. Resting-state and task-based blood oxygen level-dependent (BOLD) fMRI scans will be acquired to measure related brain function. To assess distributed and intrinsic brain functional connectivity patterns, we will acquire a resting-state fMRI scan (10 min), during which participants are asked to close their eyes, let their minds wander and not fall asleep. Participants will complete established tasks to assess processes involved in cognition and mood, e.g., the Cyberball task, a ball-tossing game during which the participant interacts with fictitious characters to simulate experiences of social inclusion, exclusion, rejection and ostracism [[68]]. Trained research personnel will instruct participants on how to perform all tasks.

PET imaging

Participants in subcohort II will undergo PET neuroimaging with [11C]-UCB-J, which binds to the presynaptic vesicle glycoprotein 2A (SV2A). SV2A is ubiquitously and homogeneously located in synapses across the brain and allows for the determination of SV2A binding and presynaptic density in the brain [[69]]. However, due to the ubiquitous distribution of SV2A, there is no proper reference region in the brain, and we, therefore, measure the arterial input function. PET scanning is conducted using a High-Resolution Research Tomography (HRRT) PET scanner (CTI/Siemens, Knoxville, TN, USA). First a 6 min transmission scan, then an intravenous bolus of < 400 MBq of [11C]-UCB-J administered over 20 s followed by a 90-min dynamic acquisition (256 × 256 × 207 voxels; 1.22 × 1.22 × 1.22 mm).

Treatment and life events

As the study is observational, we will not interfere with or delay treatment. EMR and registry data during treatment and until the 18 months from inclusion will be extracted to inform the individual treatment path, i.e., what treatment the individual patient received, e.g., amount of individual psychotherapy sessions and when, timing, type and dose of medication and switches, and participation in group therapy. Using registry data, we can also follow life events, e.g., change of residence, divorce, and employment.

Follow-up

Patient-reported outcome measures assessing depression symptom severity and clinical status are sent via the national secure mail platform at three time points: 6, 12 and 18 months after treatment start (Table 4). Complete registry follow-up is done at 18 months as well.

Table 4 Follow-up Measurements aOmitted if the patient did not receive antidepressant medication

Measures

6 months

12 months

18 months

Quick Inventory of Depressive Symptomatology (QIDS-SR)

X

X

X

BSI-18, SCL-10, WHO-5 and mSDS

X

X

X

Patient-Reported Inventory of Side-Effects (PRISE)[51]a

X

X

X

Negative Effects Questionnaire (NEQ)

X

Primary and secondary outcome measures

The primary clinical outcomes are categorical: improvement defined as ≥ 50% reduction in QIDS and remission after the treatment package defined as a QIDS ≤ 5. The secondary outcome is change in depression severity as measured by QIDS.

Tertiary outcome measures

The ten-item depression and anxiety symptom checklist (SCL-10) [[50], [71]], well-being measured by WHO-5 [[50], [72]], and disability measured by a modified Sheehan Disability Scale (mSDS) are the established treatment effect parameters by the Mental Health Services of the Capital Region of Denmark.

Tertiary endpoints are three measurements of psychosocial remission defined as a WHO-5 score of > 49, an SCL-10 score of < 26 and an mSDS score of < 10. Additional tertiary clinical endpoints are changes in wellness (WHO-5), disability (mSDS), and symptomatology on the Brief Symptom Inventory 18 (BSI-18) and the SCL-10.

Two questionnaires assessing the negative effects of psychological and antidepressant treatment will be sent at the first follow-up, at the end of the treatment package. We use the Patient-Reported Inventory of Side-Effects (PRISE), originally developed and validated in Danish and also used in the STAR*D trial [[71], [73]]. The questionnaire is omitted if the patient has not been or is not on antidepressant medication. We use the short 20-item form of the Negative Effects Questionnaire (NEQ) to assess adverse and unwanted events in psychological treatment, i.e., new symptoms, dependency, stigma, hopelessness, and the experienced quality of treatment [[74]]. Both baseline characteristics and treatment experiences, e.g., negative effects on the NEQ, will be used to investigate reasons for CBT and treatment package drop-out.

Registry follow-up

After the last 18-month follow-up, the study dataset will be sent to Statistics Denmark with a list of all invited participants to allow a non-participant analysis and long-term follow-up. The study data will be linked with data from the Danish Civil Registration System [[76]] e.g., the DNPR [[57]], the Danish National Prescription Registry [[55]], and other registries indexing, e.g., hospital admittance, diagnosis- and treatment codes, prescription medications, employment status, living situation (e.g., partner information), and income. We will also examine diagnostic stability [[77]], e.g., change of primary diagnosis from first episode depression to an anxiety or personality disorder or later recurrent depressive episode or conversion to bipolar affective disorder.

Statistical analyses

We plan to investigate the outlined hypotheses using both data- and hypothesis-driven approaches. We will publish more specific analysis plans on the web (e.g., PROSPERO) before starting detailed analyses.

Data-driven analyses

We will use machine learning algorithms to determine a combination of baseline characteristics that best predict treatment outcomes. In contrast to hypothesis-driven analyses, data-driven machine learning frameworks enable us to identify novel associations between patient characteristics and treatment response and interactive effects of complementary patient characteristic information in prediction. Recent interest in machine learning approaches to prediction is to develop classifiers that combine a range of patient-level data to provide a patient-level prediction of treatment response. The most common classifiers are based on patient characteristics or biomarkers assessed before treatment initiation [[78]]. This information may be used in clinical settings to select a specific treatment if it is predicted to have a higher chance of success or to suggest that a patient may generally be treatment-resistant, which could justify earlier use of second-line therapies [[26]].

In the present study, we will first use machine learning to train classifiers based on broad non-imaging data collected from all patients in the cohort. Neuroimaging data from EEG, MRI, and PET in subcohorts I and II will subsequently be included, enabling us to make meaningful statements about the marginal improvement in model performance with or without specific neuroimaging measures. This is critical for optimising patient care with costs associated with data acquisition. Following the recommendations for best practice [[80]], we will employ randomized k-fold nested cross-validation, i.e., splitting the collected data into training and testing datasets that reported model performance measures are not upwardly biased due to data leakage.

We will use latent class analysis to identify different MDD subgroups that share characteristics measured at baseline (hypotheses 1.2 and 2.3) [[25], [82]]. To investigate if adding neuroimaging data improves clustering (hypothesis 2.3), we will compare the heterogeneity of change on clinical outcome, i.e., mean and variance.

Hypothesis-driven analyses

To answer the hypothesis-driven research questions, i.e., hypotheses 3.1 – 3.4, we will use appropriate parametric models, including multiple linear regression or linear latent variable models to investigate group differences in [11C]-UCB-J binding between patients and HCs and multiple linear regression to determine associations between cognitive scores and [11C]-UCB-J binding in patients. We will adjust for age and sex, as our current data on HCs with [11C]-UCB-J are of equal sex distribution with a mean age of approximately 30 years old, which is not expected for the patient sample expecting to be predominately women and below 25 years old (Supplementary Fig. 1). Furthermore, dependent on additional funding, we will include more HCs and attempt to match their age distribution with the patients better.

We will also use statistical models such as logistic regression to investigate associations between individual measures and dichotomous clinical outcomes (e.g., the association between early childhood trauma and treatment response) when testing secondary hypotheses. Lastly, we will assess the associations between baseline patient characteristics, treatment events, and clinical outcomes using path analysis, enabling us to estimate the effect of treatments over time on the clinical outcome.

Power calculation

Data-driven analyses

To answer primary hypotheses 1.1 – 3.1, we will use a machine learning approach as outlined above. Statistical power calculations are not well adapted to data-driven machine learning model frameworks because any such calculation depends on broad assumptions about model structure and feature space. However, the number of patients in the cohort (N = 800) represents one of the largest cohorts to date, looking to determine prediction classifiers in MDD [[84]].

To limit the strain on patients as well as costs, we collect neuroimaging data in a subset of patients (N = 600) and not in the entire cohort. By contrast, questionnaire and EMR data collection is relatively cheap, fast, and non-invasive. Therefore, this data's potential predictive value need not be very high to be clinically relevant. Acquisition of neuroimaging data, i.e., MR and PET, is costly and time-consuming and must exhibit higher predictive value to be relevant as a clinical tool. Thus, fewer patients are needed in Cohorts II-III to determine the relevance of neuroimaging biomarkers in MDD, as the power of these biomarkers would have to be large enough to be detectable even in smaller patient samples.

Hypothesis-driven analyses

To answer hypothesis 3.1, we will use [11C]-UCB-J PET data from healthy controls (N = 40) currently available from the Cimbi database with [11C]-UCB-J PET data collected from patients in the PET subcohort II (expected n = 60). These sample sizes will provide us with a statistical power of 0.99 to detect group differences with a Cohen's d effect size of 0.95 as reported in previous study [[85]]. With the samples size and a statistical power of 0.80 we can defect a group difference with a Cohen's d 0.58 or higher using a significance threshold of p ≤ 0.05 in a two-sample t-test. The previous study by Holmes et al. (2019) reported group differences in [11C]-UCB-J binding between healthy controls and a small cohort with mixed psychiatric diagnoses, including MDD. Based on their findings in frontal cortex binding (which were similar to other brain regions), our study is statistically powered to detect group differences in binding of ~ 6.8%; notably, Holmes et al. found a group difference of 12.5% in this region, so our study should be adequately powered.

To answer hypothesis 3.2, we will have a statistical power of 0.8 to detect a significant association between [11C]-UCB-J binding and cognitive scores in the PET subcohort II equivalent to a correlation coefficient of r ≥ 0.35 at a statistical significance threshold of p ≤ 0.05.

Ethics and data availability

The study is conducted according to the principles of the seventh revision of the Declaration of Helsinki (2013) and was reviewed and approved by the Committees on Health Research Ethics in the Capital Region of Denmark (reference number: H-20083013). Before undertaking any study-related procedures, each participant receives verbal and written explanations of study aims, methods, potential hazards, and benefits from investigators and provides written informed consent. All participating patients are asked to consent to disclose relevant information from their EMR to extract health-related information relevant to the study.

Data management and monitoring during the study adhere to the rules protecting personal data. Paper-based material (e.g., cognitive test results) will be stored in a secured archive. Identifiable electronic data files will be stored in password-secured files behind a firewall per regulations.

Biological material will be coded with a unique identification number. Access to de-identification keys is restricted to authorised personnel only and stored in a temporary biobank located in secured areas in the laboratory facility. The biomaterial will later be analysed in batches to reduce noise, and potential extra material after the end of the study will be transferred to the CIMBI biobank [[32]]. All biological material will ultimately be anonymised after 15 years after the end of the study.

The study results will be presented following relevant reporting guidelines, i.e., STROBE and TRIPOD [[86]]. After publication of results from our primary hypothesis, the data can upon request be made available to other scientists or consortia, through the Cimbi database (or similar platform).

Discussion

MDD is a brain disorder with etiological heterogeneity in the interplay between biological, social, psychological, and behavioural factors and pathophysiology with several neurobiological mechanisms. In the BrainDrugs-Depression study, we employ a broad, multi-modal biopsychosocial characterisation of each patient. This, combined with a large sample size, follow-up over 18 months, and further complete follow-up in the social- and health registries, offers a unique opportunity to uncover potential single or combined predictors of treatment outcome, identify MDD subtypes, advance the understanding of MDD aetiology, and map neurobiological predictors of treatment response; ultimately paving the way for a precision medicine approach for optimised MDD treatment.

Further, the ability to track and follow patients in the Danish health care system and registers provides a unique opportunity to obtain large amounts of data collected independent of the patient's mood and enables us to perform sensitivity analyses and account for selective participation and attrition. The study also makes it possible to track the long-term consequences of various degrees of treatment response and point to areas where the current treatment could be improved to obtain better prognostic outcomes.

Cognitive dysfunction is a core dimension in MDD, both in first- and multiple-episode patients [[88]] and mediates a significant degree of psychosocial impairment and reduction in workplace productivity [[89]]. The presence of cognitive dysfunction may also significantly impact medication [[90]] and psychotherapy [[91]] and may even persist even past remission of the depressive episode [[92]]. Recently, researchers have begun to explore the potential of cognitive markers to inform clinical decision-making in the treatment of depression [[94]] and as a specific treatment target [[95]]. We include an extensive neurocognitive battery at inclusion and hope to extend the follow-up assessments to include a brief internet-based self-administered cognitive assessment [[96]] to follow long-term cognitive function.

This study is embedded in a more extensive network of closely related studies in the BrainDrugs Research Alliance (braindrugs.nru.dk), which will significantly increase its scientific scope and value. Specifically, a concurrent prospective cohort study, the BrainDrugs-Epilepsy study at our research unit, studies newly diagnosed patients with epilepsy using a similar multi-modal precision medicine approach [[33]]. Depression and other psychiatric disorders (e.g., anxiety) are frequent in patients with epilepsy, and a bidirectional relationship has been proposed [[97]–[100]]. Shared measures enable further exploration of the relationship between depression and epilepsy.

During the last decades, new developments in the pharmacological treatment of depression have been modest. Emphasis has been paid to the effect of other treatment modalities, such as psychotherapy, lifestyle modification and a combination of treatments. However, we do not know which specific groups of patients benefit from different treatment modalities. In the present project, we develop tools for prediction and uncover causal paths for treatment response in a real-world setting. Thus, the project goes beyond a traditional evaluation of existing health services with the potential to develop a more targeted treatment to be implemented and tested in the clinical setting. Furthermore, to maximise the utility of the participants' contribution, the project in also intended for cross-validating models from other research groups, data sharing and multi-centre collaborations. The datasets generated by this study will be available in the Cimbi database, which researchers can request access to [[32]].

Acknowledgements

We want to thank the participating patients, volunteers, technicians, collaboration partners, study assistants, and funding supporting this study.

Authors' contributions

GMK, MBJ, MO and VGF conceptualised the study. KHRJ wrote the manuscript together with MBJ. VNHD, MG, PMF, CI, AS, MRM, BO, MO, BWJHP, LHP, VGF, and GMK contributed to different aspects of the study design and critically revised the manuscript. All authors have approved the submitted version.

Funding

The Lundbeck Foundation (R279–2018–1145) and the Research Fund of the Mental Health Services—Capital Region of Denmark granted economic support for the study.

Availability of data and materials

The datasets generated by this study will be available in the Cimbi database. All researchers can request access to data from the Cimbi database (http://www.cimbi.dk/db).

Declarations

Ethics approval and consent to participate

The study is conducted according to the principles of the seventh revision of the Declaration of Helsinki (2013) and was reviewed and approved by the Committees on Health Research Ethics in the Capital Region of Denmark (reference number: H-20083013). Before undertaking any study-related procedures, each participant receives verbal and written explanations of study aims, methods, potential hazards, and benefits from investigators and provides written informed consent. The study will be carried out in accordance with relevant guidelines and regulations.

Consent for publication

Not applicable.

Competing interests

GMK has received honoraria as speaker for SAGE Therapeutics/Biogen and H. Lundbeck and as advisor for Sanos, and VF as a consultant for SAGE Therapeutics. MJ and VF have given talks sponsored by Lundbeck Pharma and MJ for Boehringer Ingelheim. The remaining authors declare that the research is conducted without any commercial or financial relationships that could be construed as a potential conflict of interest. MRMJ received sponsorship from Jazz Pharmaceuticals for attendance at the 14th European Epilepsy Congress.

Supplementary Information

Graph: Additional file 1: Supplementary Figure 1. The age and sex distribution of patients enteringthe treatment package during 2019-2021 before inclusion start.

Graph: Additional file 2: Supplementary Figure 2.  The annual distribution of treatment initiation at the Mental Healthcare Centres in the Capital Region of Denmark. *The Mental Health Centre Copenhagen comprises two clinics, i.e., in Frederiksberg and Nørrebro. B) The Mental Health Centres admission area and geographical locations (image made by K. R. Jensen).

Graph: Additional file 3.

Abbreviations

  • 5-HTTLPR
  • Serotonin-transporter-linked promoter region

• ABCB1

  • ATP Binding Cassette Subfamily B Member 1

• ABCC1

  • ATP Binding Cassette Subfamily C Member 1

• BDNF

  • Brain Derived Neurotrophic Factor

• BOLD

  • Based blood oxygen level-dependent

• BSI

  • Brief Symptom Inventory

• BSI-18

  • 18-Item Brief Symptom Inventory

• CAMB

  • Copenhagen Aging and Midlife Biobank

• CATS

  • Child abuse and trauma scale

• CBT

  • Cognitive behavioral therapy

• CIMBI

  • Center for Integrated Molecular Brain Imaging

• COBRA

  • Cognitive Complaints in Bipolar Disorder Rating Assessment

• COMT

  • Catechol-O-methyltransferase

• CPR

  • Centrale Personregister

• CSES

  • Coping Self-Efficacy Scale

• CSFQ

  • Changes in Sexual Functioning Questionnaire

• CVD

  • Center for Visitation og Diagnostik

• CYP2C19

  • Cytochrome P450 Family 2 Subfamily C Member 19

• CYP2D6

  • Cytochrome P450 Family 2 Subfamily D Member 6

• DAR-5

  • Dimension of Anger Reactions

• D-KEFS

  • Delis-Kaplan Executive Function System

• DNA

  • Deoxyribonucleic acid

• DNPR

  • Danish National Patient Registry

• EEG

  • Electroencephalogr

• EMR

  • Electronic medical records

• ERP

  • Event-related potentials

• ERT

  • Emotional Recognition Task

• GAD

  • Generalised Anxiety Disorder 7-item

• GP

  • General practitioner

• HBA1c

  • Hemoglobin A1C

• HC

  • Healthy control

• HRRT

  • High resolution research tomograph

• ICD

  • International Classification of Diseases

• IDS

  • Inventory of depressive symptomatology

• IMT

  • Emotional Intensity Morphing Task

• LDAEP

  • Loudness dependence of auditory evoked potentials

• LNS

  • Letter-Number Sequence

• MAAS

  • Mindful Attention Awareness Scale

• MCQ-30

  • Short form of Metacognitions Questionnaire

• MDD

  • Major Depressive Disorder

• MET

  • Moral Emotions Task

• MREG

  • Ultra-fast functional magnetic resonance encephalography

• MRI

  • Magnetic Resonance Imaging

• mSDS

  • Modified Sheehan Disability Scale

• MZQ

  • Mentalization Questionnaire

• NEQ

  • Negative Effects Questionnaire

• NRU

  • Neurobiology Research Unit

• OS-FHAM

  • Online Stimulant and Family History Assessment Module

• PAQ

  • Perth Alexithymia Questionnaire

• PBI

  • Parental Bonding Instrument

• PET

  • Positron emission tomography

• PRISE

  • Patient Reported Inventory of Side-Effects

• PSQI

  • Pittsburgh Sleep Quality Index

• PSS

  • Cohen's Perceived Stress Scale

• QIDS

  • Quick Inventory of Depressive Symptomatology

• RAVLT

  • Rey Auditory Verbal Learning Test

• RCFT

  • Rey Complex Figure Test

• RRS

  • Ruminative Reponse Scale

• S100B

  • S100 calcium-binding protein B

• SCIP-D

  • Screen for Cognitive Impairments in Psychiatry—Depression

• SCL-10

  • 10-Item depression and anxiety Symptom Checklist

• SDMT

  • Symbol Digit Modality Task

• SHAPS

  • Snaith-Hamilton Pleasure Scale

• SLE

  • Stressful life events

• SOI-R

  • Revised Sociosexual Orientation Inventory

• SRT

  • Simple Reaction Tim

• SSRI

  • Selective serotonin reuptake inhibitor

• STROBE

  • Strengthening the reporting of observational studies in epidemiology

• SV2A

  • Synaptic vesicle glycoprotein 2A

• TRIPOD

  • Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis

• TSH

  • Thyroid-stimulating hormone

• UGT1A1

  • UDP glucuronosyltransferase 1 family, polypeptide A1

• WHO

  • World Health Organization

• WHO-5

  • WHO 5 wellbeing index
Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References 1 WHO. Depression and Other Common Mental Disorders - Global Health Estimates. 2017: Geneva; World Health Organization 2 Huhn M, Tardy M, Spineli LM, Kissling W, Förstl H, Pitschel-Walz G. Efficacy of pharmacotherapy and psychotherapy for adult psychiatric disorders: a systematic overview of meta-analyses. Jama Psychiat. 2014; 71: 706-715. 10.1001/jamapsychiatry.2014.112 3 Cipriani A, Furukawa TA, Salanti G, Chaimani A, Atkinson LZ, Ogawa Y. Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: a systematic review and network meta-analysis. Lancet. 2018; 391: 1357-1366. 1:CAS:528:DC%2BC1cXjsVCrt7k%3D. 10.1016/S0140-6736(17)32802-7. 29477251. 5889788 4 Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D Report. Am J Psychiat. 2006; 163: 1905-1917. 10.1176/ajp.2006.163.11.1905. 17074942 5 Trevino K, McClintock SM, Fischer NM, Vora A, Husain MM. Defining treatment-resistant depression: a comprehensive review of the literature. Ann Clin Psychiatry Official J Am Acad Clin Psychiatrists. 2014; 26: 222-232 6 Gronemann FH, Jorgensen MB, Nordentoft M, Andersen PK, Osler M. Incidence of, risk factors for, and changes over time in treatment-resistant depression in Denmark: a register-based cohort study. J Clin Psychiatry. 2018; 79: 17m11845. 10.4088/JCP.17m11845. 29873959 7 Beshai S, Dobson KS, Bockting CLH, Quigley L. Relapse and recurrence prevention in depression: Current research and future prospects. Clin Psychol Rev. 2011; 31: 1349-1360. 10.1016/j.cpr.2011.09.003. 22020371 8 (NICE) The National Institute for Health and Care Excellence. Depression in adults: treatment and management. NG222. 2022. 9 Moriarty AS, Meader N, Snell KIE, Riley RD, Paton LW, Dawson S, et al. Predicting relapse or recurrence of depression: systematic review of prognostic models. Br J Psychiatry. 2022;221(2):448–58. Otte C, Gold SM, Penninx BW, Pariante CM, Etkin A, Fava M. Major depressive disorder. Nat Rev Dis Primers. 2016; 2: 16065. 10.1038/nrdp.2016.65. 27629598 Köhler-Forsberg K, Jorgensen A, Dam VH, Stenbæk DS, Fisher PM, Ip C-T. Predicting treatment outcome in major depressive disorder using serotonin 4 Receptor PET Brain Imaging, Functional MRI, Cognitive-, EEG-Based, and Peripheral Biomarkers: A NeuroPharm Open Label Clinical Trial Protocol. Frontiers Psychiatry. 2020; 11: 641. 10.3389/fpsyt.2020.00641 Kambeitz-Ilankovic L, Koutsouleris N, Upthegrove R. The potential of precision psychiatry: what is in reach?. Br J Psychiatry. 2022; 220: 175-178. 10.1192/bjp.2022.23. 35354501 Iniesta R, Hodgson K, Stahl D, Malki K, Maier W, Rietschel M. Antidepressant drug-specific prediction of depression treatment outcomes from genetic and clinical variables. Sci Rep-uk. 2018; 8: 5530. 10.1038/s41598-018-23584-z Dichter GS, Gibbs D, Smoski MJ. A systematic review of relations between resting-state functional-MRI and treatment response in major depressive disorder. J Affect Disorders. 2015; 172: 8-17. 10.1016/j.jad.2014.09.028. 25451389 Enneking V, Leehr EJ, Dannlowski U, Redlich R. Brain structural effects of treatments for depression and biomarkers of response: a systematic review of neuroimaging studies. Psychol Med. 2020; 50: 187-209. 10.1017/S0033291719003660. 31858931 Köhler-Forsberg K, Ozenne B, Larsen SV, Poulsen AS, Landman EB, Dam VH. Concurrent anxiety in patients with major depression and cerebral serotonin 4 receptor binding a NeuroPharm-1 study. Transl Psychiat. 2022; 12: 273. 10.1038/s41398-022-02034-5 Fisher PM, Ozenne B, Ganz M, Frokjaer VG, Dam VN, Penninx BW. Emotional faces processing in major depressive disorder and prediction of antidepressant treatment response: A NeuroPharm study. J Psychopharmacol. 2022; 36: 626-636. 1:CAS:528:DC%2BB38Xht12gurrN. 10.1177/02698811221089035. 35549538 Brandt IM, Köhler-Forsberg K, Ganz M, Ozenne B, Jorgensen MB, Poulsen A. Reward processing in major depressive disorder and prediction of treatment response – Neuropharm study. Eur Neuropsychopharm. 2021; 44: 23-33. 1:CAS:528:DC%2BB3MXotl2nsQ%3D%3D. 10.1016/j.euroneuro.2020.12.010 Widge AS, Bilge MT, Montana R, Chang W, Rodriguez CI, Deckersbach T. Electroencephalographic biomarkers for treatment response prediction in major depressive illness: a meta-analysis. Am J Psychiat. 2019; 176: 44-56. 10.1176/appi.ajp.2018.17121358. 30278789 Ip C-T, Olbrich S, Ganz M, Ozenne B, Köhler-Forsberg K, Dam VH. Pretreatment qEEG biomarkers for predicting pharmacological treatment outcome in major depressive disorder: Independent validation from the NeuroPharm study. Eur Neuropsychopharm. 2021; 49: 101-112. 1:CAS:528:DC%2BB3MXovV2kurY%3D. 10.1016/j.euroneuro.2021.03.024 Strawbridge R, Young AH, Cleare AJ. Biomarkers for depression: recent insights, current challenges and future prospects. Neuropsych Dis Treat. 2017; 13: 1245-1262. 1:CAS:528:DC%2BC1cXhsl2itr7F. 10.2147/NDT.S114542 Etkin A, Patenaude B, Song YJC, Usherwood T, Rekshan W, Schatzberg AF. A Cognitive-emotional biomarker for predicting remission with antidepressant medications: a report from the iSPOT-D Trial. Neuropsychopharmacol. 2015; 40: 1332-1342. 1:CAS:528:DC%2BC2MXhtFKktb8%3D. 10.1038/npp.2014.333 Dam VH, Stenbæk DS, Köhler-Forsberg K, Ip C, Ozenne B, Sahakian BJ, et al. Hot and cold cognitive disturbances in antidepressant-free patients with major depressive disorder: a NeuroPharm study. Psychol Med. 2021;51(14):2347–56. Kessler RC, van Loo HM, Wardenaar KJ, Bossarte RM, Brenner LA, Ebert DD. Using patient self-reports to study heterogeneity of treatment effects in major depressive disorder. Epidemiol Psych Sci. 2017; 26: 22-36. 1:STN:280:DC%2BC28nkt1ejsg%3D%3D. 10.1017/S2045796016000020 Dinga R, Marquand AF, Veltman DJ, Beekman ATF, Schoevers RA, van Hemert AM. Predicting the naturalistic course of depression from a wide range of clinical, psychological, and biological data: a machine learning approach. Transl Psychiat. 2018; 8: 241. 10.1038/s41398-018-0289-1 Gillett G, Tomlinson A, Efthimiou O. Cipriani A. 2020: A meta-review. Pharmacol Therapeut; Predicting treatment effects in unipolar depression: 107557 Beliveau V, Hedeboe E, Fisher PM, Dam VH, Jørgensen MB, Frokjaer VG. Generalizability of treatment outcome prediction in major depressive disorder using structural MRI: a Neuropharm study. Neuroimage Clin. 2022; 36: 103224. 10.1016/j.nicl.2022.103224. 36252556. 9668596 Meehan AJ, Lewis SJ, Fazel S, Fusar-Poli P, Steyerberg EW, Stahl D. Clinical prediction models in psychiatry: a systematic review of two decades of progress and challenges. Mol Psychiatr. 2022; 27: 2700-2708. 10.1038/s41380-022-01528-4 Carlo VD, Calati R, Serretti A. Socio-demographic and clinical predictors of non-response/non-remission in treatment resistant depressed patients: A systematic review. Psychiat Res. 2016; 240: 421-430. 10.1016/j.psychres.2016.04.034 Perlman K, Benrimoh D, Israel S, Rollins C, Brown E, Tunteng J-F. A systematic meta-review of predictors of antidepressant treatment outcome in major depressive disorder. J Affect Disorders. 2019; 243: 503-515. 1:CAS:528:DC%2BC1cXhvVeqsbnE. 10.1016/j.jad.2018.09.067. 30286415 Saracci R. Epidemiology in wonderland: Big Data and precision medicine. Eur J Epidemiol. 2018; 33: 245-257. 10.1007/s10654-018-0385-9. 29623670 Knudsen GM, Jensen PS, Erritzoe D, Baaré WFC, Ettrup A, Fisher PM. The Center for Integrated Molecular Brain Imaging (Cimbi) database. Neuroimage. 2016; 124: 1213-1219. 10.1016/j.neuroimage.2015.04.025. 25891375 Marstrand-Joergensen MR, Dam VH, Vinter K, Andersen NB, Ip C-T, Hoei-Hansen CE, et al. A prospective open-label cohort precision medicine study in epilepsy: BrainDrugs. Submitted to BMC Neurology. Rush AJ, Carmody T, Reimitz P. The Inventory of Depressive Symptomatology (IDS): Clinician (IDS-C) and Self-Report (IDS-SR) ratings of depressive symptoms. Int J Method Psych. 2000; 9: 45-59. 10.1002/mpr.79 Hausberg MC, Schulz H, Piegler T, Happach CG, Klöpper M, Brütt AL. Is a self-rated instrument appropriate to assess mentalization in patients with mental disorders? Development and first validation of the Mentalization Questionnaire (MZQ). Psychother Res. 2012; 22: 699-709. 10.1080/10503307.2012.709325. 22867004 Ott CV, Bjertrup AJ, Jensen JH, Ullum H, Sjælland R, Purdon SE. Screening for cognitive dysfunction in unipolar depression: Validation and evaluation of objective and subjective tools. J Affect Disorders. 2016; 190: 607-615. 10.1016/j.jad.2015.10.059. 26583350 Forbes D, Hawthorne G, Elliott P, McHugh T, Biddle D, Creamer M. A concise measure of anger in combat-related posttraumatic stress disorder. J Trauma Stress. 2004; 17: 249-256. 10.1023/B:JOTS.0000029268.22161.bd. 15253097 Griffith JW, Raes F. Factor Structure of the Ruminative Responses Scale. Eur J Psychol Assess. 2015; 31: 247-253. 10.1027/1015-5759/a000231 Sanders B, Becker-Lausen E. The measurement of psychological maltreatment: Early data on the child abuse and trauma scale. Child Abuse Neglect. 1995; 19: 315-323. 1:STN:280:DyaK2svisVKiug%3D%3D. 10.1016/S0145-2134(94)00131-6. 9278731 Spitzer RL, Kroenke K, Williams JBW, Löwe B. A Brief Measure for Assessing Generalized Anxiety Disorder: The GAD-7. Arch Intern Med. 2006; 166: 1092-1097. 10.1001/archinte.166.10.1092. 16717171 Preece D, Becerra R, Robinson K, Dandy J, Allan A. The psychometric assessment of alexithymia: Development and validation of the Perth Alexithymia Questionnaire. Pers Indiv Differ. 2018; 132: 32-44. 10.1016/j.paid.2018.05.011 Parker G, Tupling H, Brown LB. A Parental Bonding Instrument. Brit J Med Psychol. 1979; 52: 1-10. 10.1111/j.2044-8341.1979.tb02487.x Eskildsen A, Dalgaard VL, Nielsen KJ, Andersen JH, Zachariae R, Olsen LR. Cross-cultural adaptation and validation of the Danish consensus version of the 10-item Perceived Stress Scale. Scand J Work Environ Heal. 2015; 41: 486-490. 10.5271/sjweh.3510 Brown KW, Ryan RM. The Benefits of Being Present: Mindfulness and Its Role in Psychological Well-Being. J Pers Soc Psychol. 2003; 84: 822-848. 10.1037/0022-3514.84.4.822. 12703651 Kendler KS, Karkowski LM, Prescott CA. Stressful life events and major depression: risk period, long-term contextual threat, and diagnostic specificity. J Nerv Ment Dis. 1998; 186: 661-669. 1:STN:280:DyaK1M%2FjvFOksQ%3D%3D. 10.1097/00005053-199811000-00001. 9824167 Keller A, McGarvey EL, Clayton AH. Reliability and construct validity of the changes in sexual functioning questionnaire Short-Form (CSFQ-14). J Sex Marital Ther. 2006; 32: 43-52. 10.1080/00926230500232909. 16234225 Derogatis LR, Melisaratos N. The Brief Symptom Inventory: an introductory report. Psychol Med. 1983; 13: 595-605. 1:STN:280:DyaL2c%2Fhs1CnsQ%3D%3D. 10.1017/S0033291700048017. 6622612 Wells A, Cartwright-Hatton S. A short form of the metacognitions questionnaire: properties of the MCQ-30. Behav Res Ther. 2004; 42: 385-396. 10.1016/S0005-7967(03)00147-5. 14998733 Avlund K, Osler M, Mortensen EL, Christensen U, Bruunsgaard H, Holm-Pedersen P. Copenhagen Aging and Midlife Biobank (CAMB): an introduction. J Aging Health. 2014; 26: 5-20. 10.1177/0898264313509277. 24584257 Bech P, Austin SF, Lau ME. Patient reported outcome measures (PROMs): examination of the psychometric properties of two measures for burden of symptoms and quality of life in patients with depression or anxiety. Nord J Psychiat. 2018; 72: 1-7. 10.1080/08039488.2018.1451918 Chesney MA, Neilands TB, Chambers DB, Taylor JM, Folkman S. A validity and reliability study of the coping self-efficacy scale. Brit J Health Psych. 2006; 11: 421-437. 10.1348/135910705X53155 Penke L, Asendorpf JB. Beyond Global Sociosexual Orientations: A More Differentiated Look at Sociosexuality and Its Effects on Courtship and Romantic Relationships. J Pers Soc Psychol. 2008; 95: 1113-1135. 10.1037/0022-3514.95.5.1113. 18954197 Snaith RP, Hamilton M, Morley S, Humayan A, Hargreaves D, Trigwell P. A Scale for the Assessment of Hedonic Tone the Snaith-Hamilton Pleasure Scale. Brit J Psychiat. 1995; 167: 99-103. 1:STN:280:DyaK28%2FlsFOltg%3D%3D. 10.1192/bjp.167.1.99. 7551619 Buysse DJ, Reynolds CF, Monk TH, Berman SR, Kupfer DJ. The Pittsburgh sleep quality index: A new instrument for psychiatric practice and research. Psychiat Res. 1989; 28: 193-213. 1:STN:280:DyaL1Mzhs1Wmsg%3D%3D. 10.1016/0165-1781(89)90047-4 Kildemoes HW, Sørensen HT, Hallas J. The Danish National Prescription Registry. Scand J Public Healt. 2011; 39 7_suppl: 38-41. 10.1177/1403494810394717 Pottegård A, Schmidt SAJ, Wallach-Kildemoes H, Sørensen HT, Hallas J, Schmidt M. Data Resource Profile: The Danish National Prescription Registry. Int J Epidemiol. 2017;46(3):798–798f. Schmidt M, Schmidt SAJ, Sandegaard JL, Ehrenstein V, Pedersen L, Sørensen HT. The Danish national patient registry: a review of content, data quality, and research potential. Clin Epidemiology. 2015; 7: 449-490. 10.2147/CLEP.S91125 Bliddal M, Broe A, Pottegård A, Olsen J, Langhoff-Roos J. The Danish medical birth register. Eur J Epidemiol. 2018; 33: 27-36. 10.1007/s10654-018-0356-1. 29349587 Thygesen LC, Daasnes C, Thaulow I, Brønnum-Hansen H. Introduction to Danish (nationwide) registers on health and social issues: Structure, access, legislation, and archiving. Scand J Public Healt. 2011; 39 7_suppl: 12-6. 10.1177/1403494811399956 Dam VH, Thystrup CK, Jensen PS, Bland AR, Mortensen EL, Elliott R. Psychometric Properties and Validation of the EMOTICOM Test Battery in a Healthy Danish Population. Front Psychol. 2019; 10: 2660. 10.3389/fpsyg.2019.02660. 31849772. 6901831 Köhler-Forsberg O, Buttenschøn HN, Tansey KE, Maier W, Hauser J, Dernovsek MZ. Association between C-reactive protein (CRP) with depression symptom severity and specific depressive symptoms in major depression. Brain Behav Immun. 2017; 62: 344-350. 10.1016/j.bbi.2017.02.020. 28257825 Krogh J, Benros ME, Jørgensen MB, Vesterager L, Elfving B, Nordentoft M. The association between depressive symptoms, cognitive function, and inflammation in major depression. Brain Behav Immun. 2014; 35: 70-76. 1:CAS:528:DC%2BC3sXhsV2mtbzJ. 10.1016/j.bbi.2013.08.014. 24016864 Tural U, Irvin MK, Iosifescu DV. Correlation between S100B and severity of depression in MDD: A meta-analysis. World J Biological Psychiatry. 2022;23(6):456–63. Mosiołek A, Mosiołek J, Jakima S, Pięta A, Szulc A. Effects of Antidepressant Treatment on Neurotrophic Factors (BDNF and IGF-1) in Patients with Major Depressive Disorder (MDD). J Clin Medicine. 2021; 10: 3377. 10.3390/jcm10153377 Henriksen RA, Jenjaroenpun P, Sjøstrøm IB, Jensen KR, Prada-Luengo I, Wongsurawat T. Circular DNA in the human germline and its association with recombination. Mol Cell. 2021; 82: 209-217.e7. 10.1016/j.molcel.2021.11.027. 34951964 Meerman JJ, Janzing JGE, ter Hark SE, Coenen MJH. The potential of polygenic risk scores to predict antidepressant treatment response in major depression: a systematic review. J Affect Disorders. 2022; 304: 1-11. 1:CAS:528:DC%2BB38Xht1Kms7bK. 10.1016/j.jad.2022.02.015. 35151671 Hennig J, Kiviniemi V, Riemenschneider B, Barghoorn A, Akin B, Wang F. 15 Years MR-encephalography. Magnetic Reson Mater Phys Biol Med. 2021; 34: 85-108. 10.1007/s10334-020-00891-z Williams KD, Jarvis B. Cyberball: A program for use in research on interpersonal ostracism and acceptance. Behav Res Methods. 2006; 38: 174-180. 10.3758/BF03192765. 16817529 Nabulsi NB, Mercier J, Holden D, Carre S, Najafzadeh S, Vandergeten M-C. Synthesis and Preclinical Evaluation of 11C-UCB-J as a PET Tracer for Imaging the Synaptic Vesicle Glycoprotein 2A in the Brain. J Nucl Med. 2016; 57: 777-784. 1:CAS:528:DC%2BC2sXhslWgs7rI. 10.2967/jnumed.115.168179. 26848175 Finnema SJ, Nabulsi NB, Eid T, Detyniecki K, Lin S, Chen M-K. Imaging synaptic density in the living human brain. Sci Transl Med. 2016; 8: 348ra96-348ra96. 10.1126/scitranslmed.aaf6667. 27440727 Bech P, Timmerby N. An overview of which health domains to consider and when to apply them in measurement-based care for depression and anxiety disorders. Nord J Psychiat. 2018; 72: 1-7. 10.1080/08039488.2018.1465592 Topp CW, Østergaard SD, Søndergaard S, Bech P. The WHO-5 Well-Being Index: A Systematic Review of the Literature. Psychother Psychosom. 2015; 84: 167-176. 10.1159/000376585. 25831962 Clark SL, Adkins DE, Aberg K, Hettema JM, McClay JL, Souza RP. Pharmacogenomic study of side-effects for antidepressant treatment options in STAR*D. Psychol Med. 2012; 42: 1151-1162. 1:STN:280:DC%2BC38zntVShsQ%3D%3D. 10.1017/S003329171100239X. 22041458 Rozental A, Kottorp A, Boettcher J, Andersson G, Carlbring P. Negative effects of psychological treatments: an exploratory factor analysis of the negative effects questionnaire for monitoring and reporting adverse and unwanted events. PLoS One. 2016; 11: e0157503. 10.1371/journal.pone.0157503. 27331907. 4917117 Rozental A, Kottorp A, Forsström D, Månsson K, Boettcher J, Andersson G. The negative effects questionnaire: psychometric properties of an instrument for assessing negative effects in psychological treatments. Behav Cogn Psychoth. 2019; 47: 559-572. 10.1017/S1352465819000018 Schmidt M, Pedersen L, Sørensen HT. The Danish civil registration system as a tool in epidemiology. Eur J Epidemiol. 2014; 29: 541-549. 10.1007/s10654-014-9930-3. 24965263 Jørgensen TSH, Osler M, Jorgensen MB, Jorgensen A. Mapping diagnostic trajectories from the first hospital diagnosis of a psychiatric disorder: a Danish nationwide cohort study using sequence analysis. Lancet Psychiatry. 2023; 10: 12-20. 10.1016/S2215-0366(22)00367-4. 36450298 Browning M, Kingslake J, Dourish CT, Goodwin GM, Harmer CJ, Dawson GR. Predicting treatment response to antidepressant medication using early changes in emotional processing. Eur Neuropsychopharm. 2018; 29: 66-75. 10.1016/j.euroneuro.2018.11.1102 van der Vinne N, Vollebregt MA, Rush AJ, Eebes M, van Putten MJAM, Arns M. EEG biomarker informed prescription of antidepressants in MDD: a feasibility trial. Eur Neuropsychopharm. 2021; 44: 14-22. 10.1016/j.euroneuro.2020.12.005 Varoquaux G, Raamana PR, Engemann DA, Hoyos-Idrobo A, Schwartz Y, Thirion B. Assessing and tuning brain decoders: Cross-validation, caveats, and guidelines. Neuroimage. 2017; 145 Pt B: 166-79. 10.1016/j.neuroimage.2016.10.038 Poldrack RA, Huckins G, Varoquaux G. Establishment of best practices for evidence for prediction. Jama Psychiat. 2020; 77: 534-540. 10.1001/jamapsychiatry.2019.3671 Ulbricht CM, Chrysanthopoulou SA, Levin L, Lapane KL. The use of latent class analysis for identifying subtypes of depression: a systematic review. Psychiat Res. 2018; 266: 228-246. 10.1016/j.psychres.2018.03.003 Weller BE, Bowen NK, Faubert SJ. Latent class analysis: a guide to best practice. J Black Psychol. 2020; 46: 287-311. 10.1177/0095798420930932 Su C, Xu Z, Pathak J, Wang F. Deep learning in mental health outcome research: a scoping review. Transl Psychiat. 2020; 10: 116. 10.1038/s41398-020-0780-3 Holmes SE, Scheinost D, Finnema SJ, Naganawa M, Davis MT, DellaGioia N. Lower synaptic density is associated with depression severity and network alterations. Nat Commun. 2019; 10: 1529. 10.1038/s41467-019-09562-7. 30948709. 6449365 von Elm E, Altman DG, Egger M, Pocock SJ, Gøtzsche PC, Vandenbroucke JP. Strengthening the reporting of observational studies in epidemiology (STROBE) statement: guidelines for reporting observational studies. BMJ. 2007; 335: 806. 10.1136/bmj.39335.541782.AD Collins GS, Reitsma JB, Altman DG, Moons KG. Transparent reporting of a multivariable prediction model for individual prognosis or diagnosis (TRIPOD): the TRIPOD Statement. Bmc Med. 2015; 13: 1. 10.1186/s12916-014-0241-z. 25563062. 4284921 McIntyre RS, Cha DS, Soczynska JK, Woldeyohannes HO, Gallaugher LA, Kudlow P. Cognitive deficits and functional outcomes in major depressive disorder: determinants, substrates and treatment interventions. Depress Anxiety. 2013; 30: 515-527. 10.1002/da.22063. 23468126 McIntyre RS, Soczynska JZ, Woldeyohannes HO, Alsuwaidan MT, Cha DS, Carvalho AF. The impact of cognitive impairment on perceived workforce performance: Results from the International Mood Disorders Collaborative Project. Compr Psychiat. 2015; 56: 279-282. 10.1016/j.comppsych.2014.08.051. 25439523 Krueger KP, Berger BA, Felkey B. Medication adherence and persistence: A comprehensive review. Adv Ther. 2005; 22: 313-356. 10.1007/BF02850081. 16418141 Wild J, Gur RC. Verbal memory and treatment response in post-traumatic stress disorder. Brit J Psychiat. 2008; 193: 254-255. 10.1192/bjp.bp.107.045922. 18757989 Hasselbalch BJ, Knorr U, Kessing LV. Cognitive impairment in the remitted state of unipolar depressive disorder: A systematic review. J Affect Disorders. 2011; 134: 20-31. 10.1016/j.jad.2010.11.011. 21163534 Semkovska M, Quinlivan L, O'Grady T, Johnson R, Collins A, O'Connor J. Cognitive function following a major depressive episode: a systematic review and meta-analysis. Lancet Psychiatry. 2019; 6: 851-861. 10.1016/S2215-0366(19)30291-3. 31422920 Browning M, Bilderbeck AC, Dias R, Dourish CT, Kingslake J, Deckert J, et al. The clinical effectiveness of using a predictive algorithm to guide antidepressant treatment in primary care (PReDicT): an open-label, randomised controlled trial. Neuropsychopharmacol. 2021;46(7):1307–14. Dam VH, Stenbæk DS, Köhler-Forsberg K, Ip C, Ozenne B, Sahakian BJ. Evaluating cognitive disturbances as treatment target and predictor of antidepressant action in major depressive disorder: a NeuroPharm study. Transl Psychiat. 2022; 12: 468. 1:CAS:528:DC%2BB38XivVChsr3N. 10.1038/s41398-022-02240-1 Miskowiak KW, Jespersen AE, Obenhausen K, Hafiz P, Hestbæk E, Gulyas L. Internet-based cognitive assessment tool: Sensitivity and validity of a new online cognition screening tool for patients with bipolar disorder. J Affect Disorders. 2021; 289: 125-134. 1:STN:280:DC%2BB2c%2FjsVKrsg%3D%3D. 10.1016/j.jad.2021.04.039. 33979722 Kim M, Kim Y-S, Kim D-H, Yang T-W, Kwon O-Y. Major depressive disorder in epilepsy clinics: A meta-analysis. Epilepsy Behav. 2018; 84: 56-69. 10.1016/j.yebeh.2018.04.015. 29753295 Kanner AM. Depression and epilepsy: A bidirectional relation?. Epilepsia. 2011; 52: 21-27. 10.1111/j.1528-1167.2010.02907.x. 21214536 LaFrance WC, Kanner AM, Hermann B. Chapter 20 psychiatric comorbidities in epilepsy. Int Rev Neurobiol. 2008; 83: 347-83. 10.1016/S0074-7742(08)00020-2. 18929092 Bølling-Ladegaard E, Dreier JW, Kessing LV, Budtz-Jørgensen E, Lolk K, Christensen J. Directionality of the Association Between Epilepsy and Depression: A Nationwide, Register-Based Cohort Study. Neurology. 2022 https://doi.org/10.1212/WNL.0000000000201542.

By Kristian Høj Reveles Jensen; Vibeke H. Dam; Melanie Ganz; Patrick MacDonald Fisher; Cheng-Teng Ip; Anjali Sankar; Maja Rou Marstrand-Joergensen; Brice Ozenne; Merete Osler; Brenda W. J. H. Penninx; Lars H. Pinborg; Vibe Gedsø Frokjaer; Gitte Moos Knudsen and Martin Balslev Jørgensen

Reported by Author; Author; Author; Author; Author; Author; Author; Author; Author; Author; Author; Author; Author; Author

Titel:
Deep phenotyping towards precision psychiatry of first-episode depression - the Brain Drugs-Depression cohort.
Autor/in / Beteiligte Person: Jensen, KHR ; Dam, VH ; Ganz, M ; Fisher, PM ; Ip, CT ; Sankar, A ; Marstrand-Joergensen, MR ; Ozenne, B ; Osler, M ; Penninx, BWJH ; Pinborg, LH ; Frokjaer, VG ; Knudsen, GM ; Jørgensen, MB
Link:
Zeitschrift: BMC psychiatry, Jg. 23 (2023-03-09), Heft 1, S. 151
Veröffentlichung: London : BioMed Central, [2001-, 2023
Medientyp: academicJournal
ISSN: 1471-244X (electronic)
DOI: 10.1186/s12888-023-04618-x
Schlagwort:
  • Adult
  • Humans
  • Brain diagnostic imaging
  • Cohort Studies
  • Depression
  • Treatment Outcome
  • Adolescent
  • Young Adult
  • Middle Aged
  • Aged
  • Depressive Disorder, Major diagnostic imaging
  • Depressive Disorder, Major drug therapy
  • Psychiatry
Sonstiges:
  • Nachgewiesen in: MEDLINE
  • Sprachen: English
  • Publication Type: Journal Article; Research Support, Non-U.S. Gov't
  • Language: English
  • [BMC Psychiatry] 2023 Mar 09; Vol. 23 (1), pp. 151. <i>Date of Electronic Publication: </i>2023 Mar 09.
  • MeSH Terms: Depressive Disorder, Major* / diagnostic imaging ; Depressive Disorder, Major* / drug therapy ; Psychiatry* ; Adult ; Humans ; Brain / diagnostic imaging ; Cohort Studies ; Depression ; Treatment Outcome ; Adolescent ; Young Adult ; Middle Aged ; Aged
  • References: Int J Epidemiol. 2017 Jun 1;46(3):798-798f. (PMID: 27789670) ; Sci Transl Med. 2016 Jul 20;8(348):348ra96. (PMID: 27440727) ; Compr Psychiatry. 2015 Jan;56:279-82. (PMID: 25439523) ; JAMA Psychiatry. 2020 May 1;77(5):534-540. (PMID: 31774490) ; Psychol Med. 2012 Jun;42(6):1151-62. (PMID: 22041458) ; Front Psychiatry. 2020 Jul 23;11:641. (PMID: 32792991) ; Arch Intern Med. 2006 May 22;166(10):1092-7. (PMID: 16717171) ; Neuroimage. 2017 Jan 15;145(Pt B):166-179. (PMID: 27989847) ; JAMA Psychiatry. 2014 Jun;71(6):706-15. (PMID: 24789675) ; J Affect Disord. 2022 May 1;304:1-11. (PMID: 35151671) ; Psychother Res. 2012;22(6):699-709. (PMID: 22867004) ; J Affect Disord. 2016 Jan 15;190:607-615. (PMID: 26583350) ; Epidemiol Psychiatr Sci. 2017 Feb;26(1):22-36. (PMID: 26810628) ; Mol Psychiatry. 2022 Jun;27(6):2700-2708. (PMID: 35365801) ; World J Biol Psychiatry. 2022 Jul;23(6):456-463. (PMID: 34854356) ; Behav Res Ther. 2004 Apr;42(4):385-96. (PMID: 14998733) ; Psychiatry Res. 1989 May;28(2):193-213. (PMID: 2748771) ; Int Rev Neurobiol. 2008;83:347-83. (PMID: 18929092) ; J Pers Soc Psychol. 2003 Apr;84(4):822-48. (PMID: 12703651) ; Clin Psychol Rev. 2011 Dec;31(8):1349-60. (PMID: 22020371) ; Epilepsia. 2011 Jan;52 Suppl 1:21-7. (PMID: 21214536) ; Nat Rev Dis Primers. 2016 Sep 15;2:16065. (PMID: 27629598) ; Neurology. 2023 Feb 28;100(9):e932-e942. (PMID: 36414426) ; Psychother Psychosom. 2015;84(3):167-76. (PMID: 25831962) ; Psychiatry Res. 2016 Jun 30;240:421-430. (PMID: 27155594) ; PLoS One. 2016 Jun 22;11(6):e0157503. (PMID: 27331907) ; Epilepsy Behav. 2018 Jul;84:56-69. (PMID: 29753295) ; Neuroimage Clin. 2022;36:103224. (PMID: 36252556) ; Transl Psychiatry. 2022 Nov 8;12(1):468. (PMID: 36347845) ; BMC Med. 2015 Jan 06;13:1. (PMID: 25563062) ; Eur Neuropsychopharmacol. 2021 Mar;44:14-22. (PMID: 33509659) ; Mol Cell. 2022 Jan 6;82(1):209-217.e7. (PMID: 34951964) ; BMJ. 2007 Oct 20;335(7624):806-8. (PMID: 17947786) ; Transl Psychiatry. 2020 Apr 22;10(1):116. (PMID: 32532967) ; Psychol Med. 2021 Oct;51(14):2347-2356. (PMID: 32317043) ; J Clin Med. 2021 Jul 30;10(15):. (PMID: 34362162) ; Sci Rep. 2018 Apr 3;8(1):5530. (PMID: 29615645) ; Lancet Psychiatry. 2023 Jan;10(1):12-20. (PMID: 36450298) ; Eur J Epidemiol. 2018 Jan;33(1):27-36. (PMID: 29349587) ; Neuroimage. 2016 Jan 1;124(Pt B):1213-1219. (PMID: 25891375) ; Transl Psychiatry. 2022 Jul 11;12(1):273. (PMID: 35821015) ; Scand J Public Health. 2011 Jul;39(7 Suppl):12-6. (PMID: 21898916) ; J Aging Health. 2014 Feb;26(1):5-20. (PMID: 24584257) ; J Nucl Med. 2016 May;57(5):777-84. (PMID: 26848175) ; Br J Psychiatry. 2022 Aug;221(2):448-458. (PMID: 35048843) ; Adv Ther. 2005 Jul-Aug;22(4):313-56. (PMID: 16418141) ; Child Abuse Negl. 1995 Mar;19(3):315-23. (PMID: 9278731) ; Br J Psychiatry. 2008 Sep;193(3):254-5. (PMID: 18757989) ; Am J Psychiatry. 2019 Jan 1;176(1):44-56. (PMID: 30278789) ; Lancet. 2018 Apr 7;391(10128):1357-1366. (PMID: 29477251) ; Depress Anxiety. 2013 Jun;30(6):515-27. (PMID: 23468126) ; J Pers Soc Psychol. 2008 Nov;95(5):1113-35. (PMID: 18954197) ; Eur Neuropsychopharmacol. 2019 Jan;29(1):66-75. (PMID: 30473402) ; Nord J Psychiatry. 2018 May;72(4):251-258. (PMID: 29546787) ; J Affect Disord. 2011 Nov;134(1-3):20-31. (PMID: 21163534) ; Neuropsychopharmacology. 2015 May;40(6):1332-42. (PMID: 25547711) ; Transl Psychiatry. 2018 Nov 5;8(1):241. (PMID: 30397196) ; J Sex Marital Ther. 2006 Jan-Feb;32(1):43-52. (PMID: 16234225) ; J Trauma Stress. 2004 Jun;17(3):249-56. (PMID: 15253097) ; Nat Commun. 2019 Apr 4;10(1):1529. (PMID: 30948709) ; J Nerv Ment Dis. 1998 Nov;186(11):661-9. (PMID: 9824167) ; Br J Psychiatry. 2022 Apr;220(4):175-178. (PMID: 35354501) ; Behav Cogn Psychother. 2019 Sep;47(5):559-572. (PMID: 30871650) ; Neuropsychiatr Dis Treat. 2017 May 10;13:1245-1262. (PMID: 28546750) ; Eur Neuropsychopharmacol. 2021 Aug;49:101-112. (PMID: 33910154) ; Eur J Epidemiol. 2018 Mar;33(3):245-257. (PMID: 29623670) ; MAGMA. 2021 Feb;34(1):85-108. (PMID: 33079327) ; J Clin Psychiatry. 2018 May 29;79(4):. (PMID: 29873959) ; Ann Clin Psychiatry. 2014 Aug;26(3):222-32. (PMID: 25166485) ; Clin Epidemiol. 2015 Nov 17;7:449-90. (PMID: 26604824) ; Eur Neuropsychopharmacol. 2021 Mar;44:23-33. (PMID: 33455816) ; Eur J Epidemiol. 2014 Aug;29(8):541-9. (PMID: 24965263) ; Lancet Psychiatry. 2019 Oct;6(10):851-861. (PMID: 31422920) ; J Affect Disord. 2021 Jun 15;289:125-134. (PMID: 33979722) ; Behav Res Methods. 2006 Feb;38(1):174-80. (PMID: 16817529) ; Psychol Med. 2020 Jan;50(2):187-209. (PMID: 31858931) ; Psychol Med. 1983 Aug;13(3):595-605. (PMID: 6622612) ; Br J Health Psychol. 2006 Sep;11(Pt 3):421-37. (PMID: 16870053) ; Scand J Public Health. 2011 Jul;39(7 Suppl):38-41. (PMID: 21775349) ; Nord J Psychiatry. 2018 Jul;72(5):367-373. (PMID: 29714082) ; J Affect Disord. 2015 Feb 1;172:8-17. (PMID: 25451389) ; Scand J Work Environ Health. 2015 Sep 1;41(5):486-90. (PMID: 26111225) ; Psychiatry Res. 2018 Aug;266:228-246. (PMID: 29605104) ; Brain Behav Immun. 2017 May;62:344-350. (PMID: 28257825) ; Am J Psychiatry. 2006 Nov;163(11):1905-17. (PMID: 17074942) ; J Psychopharmacol. 2022 May;36(5):626-636. (PMID: 35549538) ; J Affect Disord. 2019 Jan 15;243:503-515. (PMID: 30286415) ; Br J Psychiatry. 1995 Jul;167(1):99-103. (PMID: 7551619) ; Neuropsychopharmacology. 2021 Jun;46(7):1307-1314. (PMID: 33637837) ; Pharmacol Ther. 2020 Aug;212:107557. (PMID: 32437828) ; Brain Behav Immun. 2014 Jan;35:70-6. (PMID: 24016864) ; Front Psychol. 2019 Dec 03;10:2660. (PMID: 31849772)
  • Grant Information: R279-2018-1145 Lundbeckfonden
  • Contributed Indexing: Keywords: Biomarker; Cognition; EEG; MRI; Major depressive disorder; PET; Precision medicine; Psychotherapy; SSRI; Synaptic density
  • Molecular Sequence: ClinicalTrials.gov NCT05616559
  • Entry Date(s): Date Created: 20230309 Date Completed: 20230317 Latest Revision: 20230317
  • Update Code: 20231215
  • PubMed Central ID: PMC9999625

Klicken Sie ein Format an und speichern Sie dann die Daten oder geben Sie eine Empfänger-Adresse ein und lassen Sie sich per Email zusenden.

oder
oder

Wählen Sie das für Sie passende Zitationsformat und kopieren Sie es dann in die Zwischenablage, lassen es sich per Mail zusenden oder speichern es als PDF-Datei.

oder
oder

Bitte prüfen Sie, ob die Zitation formal korrekt ist, bevor Sie sie in einer Arbeit verwenden. Benutzen Sie gegebenenfalls den "Exportieren"-Dialog, wenn Sie ein Literaturverwaltungsprogramm verwenden und die Zitat-Angaben selbst formatieren wollen.

xs 0 - 576
sm 576 - 768
md 768 - 992
lg 992 - 1200
xl 1200 - 1366
xxl 1366 -