Abstract
The field of psychiatry is hampered by a lack of robust, reliable and valid biomarkers that can aid in objectively diagnosing patients and providing individualized treatment recommendations. Here we review and critically evaluate the evidence for the most promising biomarkers in the psychiatric neuroscience literature for autism spectrum disorder, schizophrenia, anxiety disorders and post‐traumatic stress disorder, major depression and bipolar disorder, and substance use disorders. Candidate biomarkers reviewed include various neuroimaging, genetic, molecular and peripheral assays, for the purposes of determining susceptibility or presence of illness, and predicting treatment response or safety. This review highlights a critical gap in the biomarker validation process. An enormous societal investment over the past 50 years has identified numerous candidate biomarkers. However, to date, the overwhelming majority of these measures have not been proven sufficiently reliable, valid and useful to be adopted clinically. It is time to consider whether strategic investments might break this impasse, focusing on a limited number of promising candidates to advance through a process of definitive testing for a specific indication. Some promising candidates for definitive testing include the N170 signal, an event‐related brain potential measured using electroencephalography, for subgroup identification within autism spectrum disorder; striatal resting‐state functional magnetic resonance imaging (fMRI) measures, such as the striatal connectivity index (SCI) and the functional striatal abnormalities (FSA) index, for prediction of treatment response in schizophrenia; error‐related negativity (ERN), an electrophysiological index, for prediction of first onset of generalized anxiety disorder, and resting‐state and structural brain connectomic measures for prediction of treatment response in social anxiety disorder. Alternate forms of classification may be useful for conceptualizing and testing potential biomarkers. Collaborative efforts allowing the inclusion of biosystems beyond genetics and neuroimaging are needed, and online remote acquisition of selected measures in a naturalistic setting using mobile health tools may significantly advance the field. Setting specific benchmarks for well‐defined target application, along with development of appropriate funding and partnership mechanisms, would also be crucial. Finally, it should never be forgotten that, for a biomarker to be actionable, it will need to be clinically predictive at the individual level and viable in clinical settings.
Keywords: Biomarkers, neuroimaging, GWAS, treatment response, precision medicine, autism spectrum disorder, schizophrenia, depression, bipolar disorder, anxiety disorders, post‐traumatic stress disorder, substance use disorder
The search for biomarkers in psychiatry is motivated by the need for objective measures to inform diagnosis, prognosis, and treatment choices. The ultimate purpose of a biomarker is to improve management of a disease towards better outcomes 1 , allowing for preventive and therapeutic interventions that are tailored to a particular person's genes, environment and lifestyle (i.e., a precision medicine approach).
The US Food and Drug Administration (FDA) separates classes of biomarkers based on their applications 2 , and several of these are likely to impact the clinical management of mental disorders: a) susceptibility biomarkers, aimed at estimating the likelihood of developing an illness, which may inform allocation of preventive interventions; b) predictive biomarkers, aimed at estimating the likelihood of experiencing a therapeutic drug effect, which may consequently inform treatment selection; and c) safety biomarkers, aimed at predicting side effects, which may further aid in therapeutic decisions by anticipating poor tolerability.
Non‐invasive biomarkers, for example those based on magnetic resonance imaging (MRI) and electroencephalography (EEG), are of particular interest for developing personalized approaches. This is not only because they are relevant to the pathophysiology of interest, but also because it is hoped that they may be scalable and adoptable in the clinic – either now or in the near‐term future.
The general litmus test for biomarkers in psychiatric disorders is their ability to change clinical practice. To achieve this goal, several steps in their development are required.
The first stage is to identify a target clinical question that a particular biological measure may be appropriate to address. The most valuable target applications for biomarkers are those that can inform “high‐risk, high‐reward” decisions. For example, targeting decisions to prescribe a medication with potential life‐changing benefits but also serious side effects (e.g., clozapine for schizophrenia 3 ) may take priority over targeting decisions bearing less potential benefits or risks. Another relevant consideration is the extent to which a biomarker may optimize decision‐making above and beyond clinical data. In this respect, diagnostic biomarkers may be less clinically informative in cases where they are unlikely to override decisions based on patients’ complaints and clinical presentation. A final consideration is that the value of biomarkers will necessarily evolve with novel therapeutic options. For example, susceptibility biomarkers for conversion to psychosis, or for the emergence of autism spectrum disorder, would become particularly valuable in the case that interventions capable of preventing these outcomes become available.
The second stage is internal validation. In this stage, it must be demonstrated that a relevant biomarker reflects the underlying process of interest, instead of confounds or other epiphenomena. Confounds may include demographic characteristics, illness chronicity or severity, treatment, co‐occurring psychiatric and medical conditions, and site characteristics, among others. There may also be methodological confounds, such as head‐motion artifacts that are inextricably tied to the disorder or psychopathological trait itself (e.g., impulsivity 4 ), and therefore are not amenable to traditional statistical covariation 5 . Unfortunately, most biomarkers under development fail to move past the internal validation step.
The third stage is external validation. In this stage, it must be demonstrated that a biomarker has sufficient predictive validity in a sample independent from the one used to develop it. A critical impediment to external validation is overfitting. This refers to a model that excessively reflects the idiosyncratic (noisy) features of the dataset in which it is developed, so that it underperforms when applied to new data 6 . This stage of biomarker development thus focuses on minimizing overfitting and maximizing generalizability. It further focuses on considering and managing issues such as lack of diversity in clinical trials, failure to account for common comorbidities, or a potentially evolving biology over the course of a disorder. Statistical methods such as cross‐validation and resampling allow one to measure the generalizability of a model without applying it to an independent sample 7 . However, this does not replace the critical step of confirming generalizability in a fully independent sample not used for model training 6 .
At the stage of external validation, the most relevant performance metrics no longer pertain to significant statistical associations; instead, out‐of‐sample discrimination or predictive performance are most important 8 . Common metrics include the area under the curve (AUC) in receiver‐operator curves, and the hazard ratio for time‐to‐event predictions 9 . The AUC captures a trade‐off between true positives and false positives, with higher AUC values indicating improved discriminative ability to identify true positives without excessive false positives. As a general reference, the American Psychiatric Association Work Group on Neuroimaging Markers of Psychiatric Disorders suggested an AUC >0.8 as a minimally useful threshold 10 . Nonetheless, what is considered useful may at least partly depend on contextual factors such as the performance of available predictive models and the consequences of inaccurate prediction 11 , or the value of the expected information gain. For example, although available predictive models in suicide prevention have accuracy near zero 12 , incorrect predictions are catastrophic, and therefore even a marginal increase in accuracy could be highly valuable from an individual and public‐health standpoint. As a final step in external validation, calibration of trained models can be used to assess, and fine‐tune as necessary, the prediction performance across the entire range of outcome probabilities 8 .
The fourth and final stage requires demonstrating clinical utility. At this stage, biomarkers will have to exhibit added value relative to existing tools for clinical decision‐making. They must also be scalable and, ultimately, cost‐effective. This may involve model comparison against current methods of prediction, such as expert prognostication of relevant outcomes or clinical judgments 13 , 14 , in addition to chance‐level prediction. However, once again, the designation of clinical utility may be partly context‐dependent. For outcomes with especially high stakes (e.g., suicide, drug overdose, conversion to psychosis), expensive and/or marginally accurate new biomarkers may still provide high clinical value in comparison to the status quo, and ultimately may be cost‐effective if they can prevent the catastrophic outcome from occurring, particularly if they are proximal predictors of that outcome.
The goal of this paper is to describe and discuss candidate biomarkers – encompassing genetic, molecular, neuroimaging and/or peripheral assays as warranted – for autism spectrum disorder (ASD); schizophrenia spectrum disorders (hereafter referred to as schizophrenia for simplicity); anxiety disorders and post‐traumatic stress disorder (PTSD); mood disorders, encompassing major depressive disorder (MDD) and bipolar disorder (BD); and substance use disorders (SUDs). Recognizing that a listing of all potential biomarkers could be overwhelming and lack coherence, we do not provide an exhaustive list of the candidate biomarkers for each disorder that have been proposed or evaluated to date. Rather, we list and critically evaluate the evidence only for selected biomarkers which we view as especially promising for the field.
Biomarker development may be generally viewed as following a stepwise pipeline akin to that in drug development 11 . For some indications, the biomarkers reviewed here are farther along in development and closer to being clinically actionable; for other indications, the focus is on biomarkers which are earlier in development but are seen as having strong potential for breakthrough advancement once validated. We end each section with a brief summary of the reviewed literature, as well as a shortlist of what we consider to be especially promising (if applicable). These especially promising biomarkers could be prioritized for future large‐scale, highly powered studies, which in turn can provide the definitive evidence of that particular biomarker's ultimate success or failure.
BIOMARKERS IN AUTISM SPECTRUM DISORDER
Biological markers have been a focus in autism since its initial description in 1943 by L. Kanner, who noted large head size in five of eleven children 15 . Over time, research on biological markers in ASD has ranged from crude measures of head size to longitudinal imaging of the brain to sequencing of the entire genome.
Like all psychiatric diagnoses, ASD describes common behavioral features across individuals, instead of being a “disease” with a unifying pathophysiology. ASD spans a broad range of function and impairment: some patients require lifelong 1:1 care, while others are successful professionals and parents. Yet, unlike in other diagnostic categories reviewed here, ASD is a developmental disorder that presents in early childhood, with less time to identify biomarkers that predict onset, and with longitudinal outcomes typically measured in years rather than in weeks to months. Additionally, treatments are lacking for the core symptoms of autism – behavioral interventions show benefit primarily for IQ or language 16 , and medications primarily treat associated symptoms such as agitation or hyperactivity 17 .
ASD biomarkers that have been investigated to date primarily correspond to the susceptibility biomarkers discussed elsewhere in this review more than to predictive or safety biomarkers. Some have been described as stratification or subtyping biomarkers, given that they have sought to parse ASD into subgroups with common features. Truly unifying biology is expected at the level of single genes implicated in ASD, but peripheral or brain‐based biomarkers may identify larger subgroups that may predict prognosis or treatment response. Each of these approaches has some emerging data pointing to future utility, but to date only genetic testing is regularly used in the clinic.
Note that, throughout this section, we have endeavored to recognize the strong preference of many in the autistic community for identity‐first language (“autistic person”) over person‐first language (“person with ASD”) 18 , except when referring to the DSM diagnosis of ASD. This is in contrast to other mental disorders, such as SUDs, where it is suggested to avoid labeling a person by his/her disease 19 .
Genetic biomarkers
More genes are implicated in ASD than in any other DSM diagnosis. Most genetic variants are not inherited from either parent but are instead de novo mutations. These include single nucleotide variants (SNVs) and small insertions or deletions (indels) that disrupt single gene function, collectively implicating more than 100 genes to date 20 , 21 , 22 , 23 . De novo copy number variants (CNVs) are also implicated in ASD, most of which either delete or duplicate multiple genes 24 . Emerging data also point to rare inherited SNVs and CNVs that contribute to ASD risk 25 , 26 , 27 . Collectively, rare ASD‐associated SNVs and CNVs are found in about 15% of autistic individuals, although no single variant is found in more than 1%.
Rare ASD‐associated genetic variants are best conceptualized as identifying genetic syndromes within the overall population of autistic individuals. This extends our knowledge beyond syndromes that are typically identified before an ASD diagnosis, such as fragile X syndrome and tuberous sclerosis 28 . None of these rare variants leads to an ASD diagnosis in every individual, and many resulting syndromes also include dysmorphic features or involvement of other organ systems. Rare ASD‐associated variants are more often identified in individuals who also have intellectual disability (ID), but are still enriched in those without ID 29 , 30 . Evidence therefore supports genetic testing, including fragile X testing, chromosomal microarray to detect CNVs, and whole exome sequencing, for all autistic individuals 31 , 32 , although clinical uptake remains low 33 . This is unfortunate, as neurodevelopmental CNVs are enriched for congenital disorders and are correlated with multiple psychiatric and medical ailments 34 , suggesting that they could potentially be used to assess risk even beyond ASD.
Genetics‐based biomarkers could also be used to identify larger subgroups of individuals with unifying biology. As one example, the fragile X mental retardation protein (FMRP) binds to the mRNA of multiple genes implicated in ASD 23 , 29 , and individuals with disruption of any of these genes could potentially respond to a common treatment. A more concrete approach would be to cluster rare genetic variants into larger groupings that have defined impact on a signaling pathway, such as mTor signaling disinhibition in tuberous sclerosis and PTEN hamartoma syndrome 35 .
Common genetic variation may be another pathway to identifying biomarkers in ASD. The first five significant genome‐wide association studies (GWAS) loci were recently reported in ASD, presenting an opportunity to begin studying common variants that confer risk 36 . Thus far, polygenic risk scores (PRS) predict less than 3% of risk in ASD 36 , although this is likely to grow with larger GWAS sample sizes. Approaches to partitioning high and low PRS values already suggest avenues toward clinical utility in schizophrenia risk prediction or treatment response 37 , 38 , and similar opportunities may also open in ASD.
Peripheral biomarkers
Considerable effort has gone toward identifying and understanding potential peripheral biomarkers in ASD, beginning with the first description of elevated blood serotonin levels or hyperserotonemia in 1961 39 . Numerous peripheral findings have been reported in ASD blood, saliva and stool samples, including tests that have been approved for use by the FDA, but none of these has been sufficiently developed to warrant its use in the clinic. In many cases, research on peripheral biomarkers has focused on searching for correspondence to brain or behavioral features of ASD, without necessarily establishing a clear target for the biomarker's clinical utility, such as prediction of diagnosis or response to treatment.
The serotonin system provides an instructive example of the approaches taken to peripheral biomarkers in ASD. The description of hyperserotonemia was an early indicator of a biological origin for ASD 39 , in contrast to early attribution of the condition to parenting style 40 . Even while the diagnosis of ASD has climbed from very rare to about 2% of school‐aged children 41 , rates of hyperserotonemia (>95th percentile) have remained stable at more than 25% in ASD (meta‐analysis p=10−12) 42 . Various approaches to validation have been applied, including demonstration that hyperserotonemia is specific to ASD 43 , heritable 44 , primarily seen in boys 45 , and more commonly seen in families with multiple affected children 46 , but not associated with a particular clinical pattern 47 . Despite investigations spanning six decades, no prospective study has yet assessed whether hyperserotonemia may predict ASD risk in infants or whether it may predict treatment response to medications that target the serotonin system 47 .
Numerous other candidate peripheral biomarkers have been identified, although with less consistency across studies or specificity to ASD. Elevated pro‐inflammatory cytokines, particularly interleukin‐6 (IL‐6) and IL‐1β, have been described in several studies and are supported by meta‐analysis 48 . Increased markers of oxidative stress have also been reported, again supported by meta‐analysis 49 . Multiple groups have found differences in components of the stool microbiome in ASD, with some support but also inconsistency noted in meta‐analysis 50 .
Overall, these peripheral studies have identified broad patterns of difference between groups of autistic children or adults and comparison groups, but have not evaluated their utility as clinical biomarkers. As noted in other sections below, these markers may be largely non‐specific, due to overlap with other psychiatric and medical conditions. One group, though, has been systematically evaluating folate receptor‐α autoantibody as a potential biomarker in relation to treatment 51 . Initial data are intriguing, indicating that those with the antibody are more likely to show improved verbal communication following folinic acid administration in a placebo‐controlled pilot trial 52 . External validation of the biomarker and direct replication of these effects are still needed.
Finally, some groups have focused on composite biomarkers, including transcriptome and metabolome profiles, primarily focused on predicting ASD diagnosis. Initial studies of 100‐200 participants provided some hope that lymphocyte transcriptome profiles might separate autistic children from typically developing controls, but lacked a prospective approach 53 and/or were not specific to ASD versus developmental delay 54 . An industry‐funded study of 880 participants failed to find any transcriptome or metabolome signature with potential clinical utility in predicting diagnosis in preschoolers recruited prior to ASD evaluation (NCT01810341). In contrast, an industry‐funded study of 708 preschoolers with ASD versus non‐referred controls reported a cluster of “metabotypes” that predicted ASD diagnosis with a sensitivity of 53% and specificity of 91% 55 , with 17% having a branch chain amino acid profile with higher sensitivity. This NeuroPointDx ASD Test is currently marketed to consumers without any evidence that it prospectively improves ASD screening or diagnosis, or that it is useful to guide potential treatment. This marketing of a test without requiring FDA approval or prospective evaluation is a cautionary tale for clinician‐scientists collaborating with industry to test biomarkers in ASD.
Central nervous system biomarkers
Researchers have sought a brain signature of ASD since the advent of neuroimaging. In the last three decades, many studies have focused on potential brain‐based biomarkers quantified by a variety of indicators and techniques. These include head size as a proxy for brain size, structural MRI (sMRI) to delineate the morphology of brain structures, functional MRI (fMRI) and EEG to elucidate brain function, and cerebrospinal fluid (CSF) sampling as a measure of brain neurochemistry.
Kanner's original description of autism noted macrocephaly in some but not all cases. Consistent with this, later systematic examinations of head size in autism found macrocephaly in a subgroup that showed increased head growth after birth through early childhood 15 , 56 . Subsequent work suggested an initial surge in head growth in infants followed by a regression of growth in late childhood, but methodological problems weaken these results 57 . A recent longitudinal study confirmed the initial observation: a subgroup of ~15% showed persistent macrocephaly, primarily driven by gray matter and cortical surface area, whereas the rest of the ASD sample showed no difference from the control population 58 . Further, those with macrocephaly showed more cognitive impairment and less improvement over time 59 .
While the early observation of macrocephaly pointed to the origins of ASD in the brain, structural neuroimaging studies have not consistently found particular brain regions to be implicated in ASD 60 , 61 . Resting‐state functional connectivity studies have found complex patterns of altered connectivity in ASD, with evidence for both over‐connectivity and under‐connectivity in short‐ and long‐range networks 62 , 63 . Most promising as potential biomarkers are the findings of longitudinal neuroimaging studies in infants who have an older sibling with autism (“baby siblings”) and are therefore at elevated familial risk. In this population, changes in gray matter growth and white matter connectivity across a child's first 6‐24 months show robust prediction of later ASD diagnosis 64 , 65 . These studies have also found increased extra‐axial fluid volume in babies and toddlers who are later diagnosed with ASD 66 , 67 , and in toddlers after diagnosis 68 .
Only a minority of autistic children can tolerate an MRI scan, and EEG approaches may offer a more feasible alternative. Like MRI measures, EEG results suggest diminished long‐range connectivity, but there is inconsistency across studies 69 , 70 . EEG offers the benefit of low cost and high temporal resolution, despite poor spatial resolution. Investigators use event‐related potentials (ERPs) to evaluate processing of sensory stimuli, including social cues. The ERP response to faces is particularly characteristic, with a negative deflection at approximately 170 milliseconds (N170) showing a delay in many autistic children 71 , 72 . This N170 signal has been well replicated and validated across multiple groups, and is the only ASD biomarker to date to be submitted to the FDA. The initial target of the FDA submission is subgroup identification within ASD, but there may be future potential as a marker of treatment response as well 73 , 74 .
Neurochemical markers have also generated considerable interest in ASD. Magnetic resonance spectroscopy (MRS) studies have suggested possible regional changes in GABA or glutamate levels, though findings are inconclusive 75 , 76 . Positron emission tomography (PET) and single photon emission computed tomography (SPECT) studies have indicated decreased serotonin receptor 5‐HT2A binding 77 , which could inform treatment studies using medications that block 5‐HT2A in addition to other receptors.
Recently, decreased CSF vasopressin in neonates has been associated with later ASD diagnosis 78 . Parallel findings indicate an association between CSF vasopressin levels and symptom severity in ASD 79 . Following an initial pilot study with promising results for intranasal vasopressin in ASD 80 , it would be logical to assess CSF vasopressin as a potential biomarker of treatment response.
Finally, eye‐tracking is sometimes described as a biomarker in ASD. However, most eye‐tracking studies represent a fine‐grained analysis of behavior, rather than a biomarker per se – except perhaps for pupillometry, which is occasionally applied. Non‐biased approaches to behavioral observation are quite promising in ASD, but are beyond the scope of this review.
Summary of autism biomarkers
Biomarker development in ASD has rarely been systematic, but several potential biomarkers hold promise for future studies.
Genetic testing is now recommended for every child with an ASD diagnosis, with findings identifying genetic syndromes that often explain most of a child's risk, but are not specific to ASD. Some peripheral findings are well replicated, but most have not been assessed prospectively and none has been adequately tested for clinical utility.
Brain‐based markers show promise for subgrouping individuals, and there is some initial evidence in baby sibling studies showing that longitudinal neuroimaging can provide neural signatures that precede ASD diagnosis. After diagnosis, the EEG/ERP N170 signal has been most rigorously tested as a biomarker, with promise for identifying a subgroup within ASD and some potential as an indicator of treatment response. Finally, signals across domains support further study of serotonin‐ and vasopressin‐based biomarkers in relation to subgrouping or response to targeted treatment.
BIOMARKERS IN SCHIZOPHRENIA
The specific relevance of biomarkers for schizophrenia lies in the large burden related to this disease 81 and the costly consequences of trial‐and‐error approaches to clinical decision‐making. Delays in effective treatment involving repeated failed trials unnecessarily prolong social impairment and personal suffering, and can increase danger to self or others. Furthermore, multiple failed trials can undermine treatment engagement, which is generally already tenuous in schizophrenia, especially in the early phases of the illness 82 .
Schizophrenia is the psychiatric diagnosis with the most research on personalized biomarker approaches after depression 83 . Recent papers have provided a broad overview of biomarkers for this disorder 84 , 85 , including target biomarkers for drug development 2 , 86 and diagnostic biomarkers for pathophysiological interrogation 84 . This section will mainly focus on candidate neuroimaging biomarkers that have shown potential for eventual clinical applications by virtue of their ability to allow out‐of‐sample predictions at the individual‐subject level (i.e., beyond in‐sample statistical associations at the group level). Discussed topics include prediction of conversion to psychosis, treatment response, treatment discontinuation, and relapse risk, among others. We will, however, make exceptions in the case of relevant potential applications for which predictive performance has not yet been evaluated. In such cases, we will discuss statistical associations as examples of the preliminary stages of biomarker development.
Susceptibility biomarkers of conversion to psychosis
Susceptibility biomarkers to estimate the risk of conversion to psychosis at the individual level could be highly useful in many ways. They could indicate which subjects at clinical high risk (CHR) for psychosis are most likely to develop a full‐blown psychotic disorder, which could motivate earlier initiation of available treatments 87 , 88 to reduce the duration of untreated psychosis and its associated impact. Prognosis associated with these biomarkers would also have inherent value in preparing patients and families for what to expect in terms of chronicity and prognosis. Finally, susceptibility biomarkers could facilitate personalized treatment selection of novel disease‐ modifying agents as they become available 89 , 90 , 91 .
Prognostic models based solely on clinical data are reasonably developed. For example, the North American Prodrome Longitudinal Study (NAPLS2) individualized risk calculator 92 predicted conversion to psychosis with an AUC of 0.71 in the development cohort, and subsequently was externally validated in two large independent cohorts with an AUC ranging between 0.63 and 0.79 93 , 94 . A machine‐learning model from the Personalized Prognostic Tools for Early Psychosis Management (PRONIA) consortium achieved substantial prognostic accuracy using only clinical data, showing a balanced accuracy of 76.9% 14 . Similar predictive accuracy was achieved with the Columbia risk calculator (i.e., 73%), based on data from the Structured Interview for Prodromal Syndromes 95 . A major challenge for clinical predictive models is that they are unlikely to modify clinical practice, as high‐scoring individuals will have greater symptom burden and may already be allocated additional resources.
Neural susceptibility biomarkers could be clinically useful if they improve predictions above and beyond what is possible using clinical data. Investigators from the PRONIA consortium trained machine‐learning algorithms using clinical and gray matter volume maps to predict impaired function in a CHR cohort of 116 individuals, of whom 66 met impairment criteria at one‐year follow‐up 14 . The model using clinical data predicted social function at outcome with a balanced accuracy of 76.9%, which improved to 82.7% when adding volumetric MRI data.
In another study, the same group optimized a predictive algorithm for conversion risk in CHR states by sequentially integrating clinical‐neurocognitive‐based, expert‐based, PRS‐based, and sMRI‐based risk estimates for individual subjects, which resulted in a combined balanced accuracy of 85.9% (84.6% sensitivity, 87.3% specificity) using leave‐one‐site‐out cross‐validation 96 . The accuracy for the algorithm combining risk estimates across modalities surpassed that based solely on clinical and neurocognitive data or other individual modalities. Notably, this stepwise algorithm only required additional modalities that were deemed necessary (i.e., MRI data would be required only if clinical data were insufficient), a feature that could improve the feasibility of its clinical implementation by reducing costs and diagnostic burden.
In a smaller study, striatal glutamate measured by MRS showed promise in 19 individuals, including 7 converters. The accuracy of a predictive model based on clinical information alone was 82.1%, and this increased to 86.9% when adding a striatal glutamate measure into the model 97 .
Other biological measures that could be integrated in predictive algorithms show some promise. One example is a neuroanatomical‐maturity marker developed by the NAPLS2 consortium: in secondary analyses of 275 CHR adolescents (39 converters), a “brain‐age‐gap” marker showed an AUC of 0.63 in predicting conversion in the development sample using 10‐fold cross‐validation 98 . Another promising candidate is the EEG‐based mismatch negativity (MMN): in a study of 62 research participants who were categorized into high or low risk based on MMN, the respective hazard rate for conversion was 85% versus 13% 99 .
These and other encouraging results showing in‐sample associations 100 , 101 call for additional studies directly testing the predictive ability and generalizability of MMN as a susceptibility biomarker for conversion. Similarly, while resting‐state fMRI measures, including indices of cerebello‐thalamo‐cortical connectivity 102 , have shown robust results in association studies, their ability to improve upon the predictive capabilities of structural, neurochemical or MMN‐based measures – alone or in combination – remains to be studied. Overall, though, most susceptibility neural biomarkers for conversion have not yet been tested against models using clinical information.
It is important to note that the candidate markers reviewed here have undergone varying levels of validation. For instance, the volumetric MRI model from PRONIA 14 showed internal validity in several analyses, ruling out scanner/site effects by using nested leave‐one‐site‐out cross‐validation and assessing various effects of site, image quality, follow‐up interval, and baseline social function. Similar internal‐validation steps were taken for the NAPLS2 brain‐age‐gap marker 98 , but not for the striatal glutamate biomarker 97 . With respect to external validation, the initial gray‐matter‐volume PRONIA biomarker 14 used rigorous cross‐validation but so far lacks validation in independent samples. In contrast, the subsequent multimodal algorithm for conversion risk from PRONIA was externally validated in independent samples, yielding a balanced accuracy of 65.3‐70.4%. This exception withstanding, lack of independent validation remains a caveat that applies to most candidate biomarkers.
Finally, striatal dopamine excess has been measured with PET in CHR subjects 103 , 104 . Elevated striatal [ 18 F]DOPA uptake precedes the onset of psychosis 105 , correlates with greater severity of prodromal symptoms and neuropsychological impairment, predicts conversion, and, in both the prodrome and schizophrenia, relates negatively to prefrontal cortical activation during cognitive tasks 106 , 107 , although findings are not consistent 108 , 109 . Uptake is also predominant in the associative striatum 110 , 111 . Thus, striatal [ 18 F]DOPA uptake could advance to validation in multisite studies as a predictive diagnostic biomarker, but the cost and limited availability of PET facilities may limit its practicality. Neuromelanin‐sensitive MRI 112 , a non‐invasive and reliable measure of nigrostriatal dopamine function relevant to psychosis 113 , could provide an alternative. However, more work is needed to show its potential and justify the investment in broader testing and validation.
Susceptibility biomarkers of complications in psychosis
Susceptibility biomarkers could be clinically useful if they predict the development of complications over the course of schizophrenia. One example is the prediction of violent and/or self‐injurious behavior, for which individuals with schizophrenia are at risk 114 , 115 . Some models using clinical information have shown fair in‐sample associations 116 , 117 , but their generalizability needs to be evaluated. Neuroimaging studies have similarly shown associations of certain structural and functional features with dangerous behaviors 118 , and in some cases these neuroimaging measures have been studied alongside clinical variables 119 . Further biomarker development is needed in this area.
Predictive biomarkers of treatment response
Acute psychosis
Around 20‐30% of individuals may have treatment‐resistant schizophrenia 120 , and for these patients clozapine is the only approved drug 39 , 40 , 120 , 121 . Despite its clear superiority over other antipsychotics, clozapine has response rates neighboring 40% and carries potentially life‐threatening side effects 3 . Thus, expediting clozapine treatment for those likely to exhibit treatment resistance and who may benefit most from clozapine represents a relevant target for predictive biomarkers. More broadly, biomarkers of treatment response could further aid in personalized treatment selection, as more treatments with distinct mechanisms of action, such as cholinergic agents 122 , become available.
Perhaps the most advanced treatment‐response biomarkers along the development pipeline focus on striatal resting‐state fMRI measures, including the functional striatal abnormalities (FSA) index 123 and the striatal connectivity index (SCI) 124 .
The FSA index has been conceptualized as a diagnostic classifier. It was developed using data from 1,100 participants across seven sites, incorporating measurements that included fractional amplitude of resting‐state derived low‐frequency fluctuations 125 of striatal voxels as well as intra‐ and extra‐striatal functional connectivity of those voxels. A support vector machine (SVM) classifier was first trained to predict the diagnostic status of each individual using these features, and the FSA for any given individual was defined as the distance in the SVM feature space to the hyperplane separating cases and controls. Using leave‐one‐site‐out cross‐validation, the FSA discriminated cases from controls with 80% accuracy 123 .
In a second step, the investigators measured the association between FSA scores and symptom change over six weeks of antipsychotic treatment in a subset of 91 individuals from two of the sites. At both sites, more control‐like FSA scores showed moderate‐to‐strong correlations with improvements in total symptom severity (r=0.62, p<0.001 and r=0.42, p<0.001, respectively). Substantial efforts were made to rule out confounding factors associated with antipsychotic treatment, head motion, baseline symptom severity, and site effects, and to show specificity of the FSA relative to other psychiatric diagnoses and markers based on non‐striatal fMRI features 123 . However, a caveat is that the FSA's predictive ability was not externally validated, as the demonstration of treatment‐response prediction relied on data trained to classify diagnosis (although in orthogonal tests).
The SCI was first developed on a cohort of 41 individuals undergoing initial treatment with aripiprazole or risperidone, 24 of whom responded to treatment over 12 weeks. Functional connectivity maps for six bilateral subregions within the striatum 126 were used to identify pairwise connectivity features associated with time to response in univariate tests. Ninety‐one features were identified and weighted according to their association with time to response, and this information was used to estimate a scalar value summarizing the connectivity profiles for each individual, albeit using univariate methods without cross‐validation. Nonetheless, external validation was demonstrated in an independent cohort of 40 individuals with multi‐episode schizophrenia, where individual SCI scores, calculated with the same methodology as the discovery cohort, discriminated between responders and non‐responders with an AUC of 0.78 126 . Further studies have linked the SCI to processes implicated in treatment responsiveness, such as duration of untreated psychosis 127 , relapse 128 , and interactions with cannabis use 129 , providing some support for its internal validity.
Non‐striatal functional biomarkers have also been examined. Resting‐state functional connectivity between bilateral superior temporal cortex and other cortical regions was used in an SVM classifier model to predict 10‐week risperidone response in 38 medication‐naïve individuals 130 . This classifier showed a leave‐one‐subject‐out cross‐validation accuracy of 82.5%, although checks of internal validity were limited. Other studies have tested measures of hippocampal functional connectivity 131 , 132 , or anterior cingulate cortex glutamate levels measured with spectroscopy 133 , 134 , 135 , 136 , as potential biomarkers of treatment response, but so far these studies have only provided support for in‐sample statistical associations and are awaiting further validation. This is also the case for neuromelanin‐sensitive MRI 112 , which is currently being tested 137 as a potential biomarker of treatment response, building on previous dopamine PET work 138 .
While elevated dopamine levels in the striatum are linked to antipsychotic treatment responsiveness 139 , 140 , no prospective studies have assessed their potential utility as a biomarker for treatment response. This relates to the difficulty in implementing PET biomarkers at large scale and the limited therapeutic choices from which to select, in the case that patients were found to be non‐responsive to currently used antipsychotics, which are all acting on D2 receptors.
Psychosis relapse
Another major problem in schizophrenia is the frequency and burden of relapse, underscoring the need for predictive biomarkers of this important clinical outcome. About 80% of patients with schizophrenia will relapse at least once over the course of their illness, and many of these patients will relapse numerous times 141 , 142 . Relapse correlates to increased potential danger to self or others, and to cumulative decrements in treatment responsiveness 142 , 143 .
Antipsychotic drugs, in addition to mitigating acute psychotic symptoms, are efficacious in preventing relapse 144 . So, relapse usually occurs after interruption of maintenance antipsychotic treatment 145 , although many patients relapse while on maintenance treatment 146 . Because pathophysiological differences may exist between relapse that occurs while patients are on or off antipsychotics, these two scenarios warrant separate lines of investigation 147 . Biomarkers could allow identification of low‐relapse‐risk individuals as candidates for monitored interruption of maintenance treatment, and for high‐relapse‐risk individuals as requiring more intensive and consistent intervention 148 . To date, the literature on this topic is fairly limited, and no validated biomarkers have shown predictive value as yet. Nonetheless, exploratory analyses suggest that the SCI may be distinctly sensitive to relapse associated with treatment interruption 128 , which encourages its study in the future as a potential biomarker of relapse risk, in addition to its utility in diagnostic prediction.
Cognitive dysfunction
Although most biomarkers to date have aimed to predict and intervene on positive symptoms of schizophrenia, interest has recently emerged in developing predictive biomarkers for interventions addressing cognitive dysfunction. MMN has been studied for this purpose, based on its favorable test‐retest reliability 149 , 150 and the relationship between its deficits and cognitive dysfunction in schizophrenia 149 , 150 , 151 .
To date, no investigations have reported cross‐validated performance of MMN in predicting treatment response to cognitive training, but several studies are suggestive in this respect. For example, one trial found that the change in MMN after 1 hour of auditory cognitive training was associated with the final improvements seen with a full course of treatment 152 . Two other studies found that baseline MMN deficits predicted greater gains across various cognitive domains in response to a similar type of training 153 , 154 . Thus, converging data suggest the potential for developing MMN as a predictive biomarker for treatments addressing cognition. However, internal validity remains to be established, particularly as MMN may reflect illness duration and nicotine smoking 155 . Moreover, utility for this class of biomarkers will ultimately depend on the availability of effective interventions for cognitive dysfunction in schizophrenia.
Other pragmatic outcomes
A critical aspect of successful biomarker development is real‐world implementation. For example, the SCI was related to length of hospital stay 123 , speaking to its potential impact in real‐world clinical settings. Other early examples include a study predicting treatment discontinuation on the basis of subfield hippocampal volumes 156 , which reported that dentate gyrus volume predicted treatment disengagement with an AUC of 0.75.
However, as real‐world implementation must follow other necessary steps in the biomarker development pipeline, most candidate biomarkers are not ready for testing in real‐world clinical settings.
Predictive biomarkers of medication side effects
The prediction of treatment side effects, while highly important, is likely among the most challenging goals for neurophysiological biomarkers in schizophrenia. This is because some side effects may be partly due to non‐neural mechanisms (e.g., atypical antipsychotics modulating insulin effects on adipocytes 157 ), and some of the most serious side effects, such as neuroleptic malignant syndrome 158 , are relatively rare.
Bearing in mind these limitations, neural markers for weight gain have been studied based on observations that atypical antipsychotics can enhance anticipatory reward activations to food in the striatum 159 and affect the hypothalamic histaminergic system 160 . Some initial data suggest that striatal function may predict antipsychotic‐induced weight gain. For example, in a trial with amisulpride in 69 early‐phase patients, weight gain was associated with low reward‐ related activation of the right putamen 161 . In another clinical trial in 81 early‐phase patients treated with atypical antipsychotics, baseline left putamen volume and lower sensory‐motor connectivity at rest correlated with weight gain 162 .
This area of biomarker development is thus at a preliminary stage and requires more work. Similarly, other side effects for which neural measures may be appropriate, such as tardive dyskinesia 163 , represent a potential target for future development.
Summary of schizophrenia biomarkers
Recent efforts are advancing biomarker development for schizophrenia, particularly in clinically relevant areas of prediction of conversion to psychosis in at‐risk individuals and prediction of treatment response in acute psychosis.
In the area of conversion prediction, we have discussed examples of well‐validated multimodal algorithms incorporating sMRI and clinical information (and in some cases genetic and other data), which have reached the third stage of external validation in the biomarker development pipeline. In the area of treatment‐response prediction, we have also discussed reasonably well‐developed candidate biomarkers, mostly those based on striatal resting‐state fMRI (FSA 123 and SCI 124 ), which show different levels of generalizability in external samples.
While none of the reviewed candidate biomarkers have established clinical utility – the fourth and final stage of biomarker development required for incorporation into clinical practice – the highlighted candidates are cause for optimism. For these most promising candidates, definitive demonstrations of external validity by independent groups or via large‐scale international studies are recommended, followed by demonstrations of clinical utility. Investing in these studies, particularly those using relatively accessible measures (e.g., sMRI, PRS, resting‐state fMRI) for clinically actionable indications (e.g., conversion and treatment‐response prediction) seems well justified, given supportive evidence and high potential impact on clinical practice. Further development of multimodal sequential algorithmic workflows with the ability to decrease costs and diagnostic burden 96 also seems a fruitful area for future work. Finally, since there is no guarantee that these highlighted candidate biomarkers will show clinical utility, development of other reliable, broadly accessible, and well‐motivated measures (e.g., EEG MMN and neuromelanin‐sensitive MRI 112 ) currently at the early stages is still warranted.
A potential genetic biomarker is the PRS derived from the latest GWAS in schizophrenia, which can index substantial differences in liability between individuals. Compared with the lowest centile of PRS, the highest centile of PRS has an odds ratio for schizophrenia of 39 (95% CI: 29‐53). However, the clinical utility of the PRS as a diagnostic biomarker is limited, since the median area under the receiver operating characteristic curve (AUROC) is only 0.72, meaning that the liability explained is insufficient for predicting diagnosis in the general population 164 .
BIOMARKERS IN ANXIETY DISORDERS
Anxiety disorders – encompassing specific phobias, social anxiety disorder, agoraphobia, panic disorder, generalized anxiety disorder (GAD), separation anxiety disorder, and selective mutism – are the most frequent mental disorders, with a 12‐month prevalence of 10‐14% 165 , 166 , 167 . These disorders generate a substantial socioeconomic burden 168 , as well as significant direct and indirect health care costs 169 . They are also highly comorbid with each other, and carry an increased risk of sequential comorbidity with depression 170 and SUDs 171 .
Cognitive‐behavioral psychotherapy (CBT) and various psychotropic medications have proven efficacy in anxiety disorders, but treatment response is achieved in only half to two thirds of cases 172 , 173 , 174 . Accordingly, these disorders often follow a chronic course, with a high rate of recurrence (32.1%), and may even show stable treatment resistance (8.6%) at nine‐year follow‐up 175 .
Given this high burden and limited treatment efficacy, the identification of valid biomarkers for anxiety disorders is critical. Multiple biological mechanisms that may potentially serve as biomarkers of pathogenesis or treatment response to psychotherapy or pharmacotherapy have been identified 176 , 177 , 178 , 179 , 180 , 181 . Selected findings based on genetic, neuroimaging, neurochemical, neurophysiological and/or neurocognitive assays, which could potentially lead to biomarkers with additional validation, are presented below.
Susceptibility biomarkers
Genetics
Candidate gene studies have found that COMT (rs4680, G [val] allele), NPSR1 (rs324981, T allele), TPH1 (rs1800532, AA genotype), HTR2A (rs6313, T allele), and MAOA (uVNTR, long alleles) gene variants are most consistently involved in the pathogenesis of panic disorder. OXTR (e.g., rs2254298 GG genotype), SLC6A4 (5‐HTTLPR, short [s] allele), MAOA (uVNTR, long alleles), and HTR1A (rs6295, G allele) gene variation is most consistently involved in other anxiety phenotypes 182 , 183 , 184 , 185 . However, since replication has largely been elusive, candidate gene studies have mostly given way to GWAS, a more powerful and unbiased approach.
GWAS conducted in cooperative efforts – such as the ANGST (Anxiety NeuroGenetics Study) consortium, the UK Biobank, and the Danish iPSYCH study – have suggested that several single nucleotide polymorphisms (SNPs) in genes such as ESR1, GLRB, MYH15, NTRK2, PDE4B, RBFOX1, SATB1, TMEM132D, TMEM106B, and a non‐coding RNA locus associated with the CAMKMT gene, are linked to anxiety‐related traits, current anxiety symptoms, or lifetime anxiety disorders 186 . The largest GWAS of anxiety traits to date, using the Million Veteran Program dataset, identified genome‐wide significant associations with the Generalized Anxiety Disorder 2‐item (GAD‐2) score near genes involved in global regulation of gene expression (SATB1) and the estrogen receptor alpha (ESR1) 187 . These are promising leads, but the effects require confirmation as more data are acquired.
Given the close interaction of genetic factors with environmental influences in the pathogenesis of anxiety disorders, studies are increasingly testing gene‐environment (GxE) interaction effects using both candidate gene (e.g., RGS2 188 ) and genome‐widee.g., 189 approaches. These, too, have produced initial, but as‐yet unreplicated results. The GxE concept has recently been expanded to include a dimension of “coping”, yielding a three‐dimensional model (GxExC). For instance, replicated evidence has been reported for an interactive effect on trait anxiety of a neuropeptide S receptor (NPSR1) gene variant, early adversity, and coping factors, such that adaptive coping compensates for the otherwise deleterious effects of a GxE risk constellation 190 .
Finally, growing evidence is emerging for epigenetic mechanisms that can bridge between genetic and environmental levels 191 . For instance, altered DNA methylation patterns in the MAOA, OXTR, BDNF, NET, GAD1, CRHR1 and NR3C1 genes have been associated with panic disorder or social anxiety disorder 192 , 193 . In addition, still‐underpowered epigenome‐wide association studies (EWAS) in panic disorder and social anxiety disorder suggestively point to altered DNA methylation in previously unidentified risk genes 194 , 195 , 196 , 197 .
Given the small effect sizes of individual genetic variants, a combination of genetic, epigenetic and further molecular markers might be more informative than genetic or epigenetic data alone. For example, as the field matures, PRS such as those used to predict illness course in schizophrenia 198 could be developed for anxiety disorders. An important caveat in epigenetic studies is that they have largely relied on peripheral tissues (with the exception of investigations in post‐mortem samples), and epigenetic changes in peripheral tissues do not provide direct evidence of changes in the central nervous system 199 . Providing some optimism, though, studies that compared peripheral and central tissue have often reported considerable functional overlap 192 .
Neuroimaging
Numerous structural and task‐related fMRI, PET, SPECT and MRS studies have been conducted in anxiety disorders. Structural studies have not provided consistent results, in terms of regions or directionality. In functional studies, altered brain activation elicited in response to words or pictures with anxiety‐ or fear‐related content has been observed in the “fear network”. This includes both increased and decreased inhibitory control‐relevant activity in the orbitofrontal and in the dorsolateral, dorsomedial and ventrolateral prefrontal cortex, as well as mostly increased activity in limbic structures such as the amygdala, insula, anterior cingulate cortex, bed nucleus of the stria terminalis (BNST), and striatum. The BNST may be involved in sustained rather than phasic anxiety 180 , 200 .
Particularly consistent evidence has emerged for increased amygdala reactivity towards negative emotional stimuli in combination with insufficient prefrontal control 201 . One interpretative constraint, though, is that this activation phenotype tends to be seen across anxiety, stress‐related and mood disorders, limiting its value as a biomarker able to distinguish between clinical presentations.
Psychophysiological assays and challenges
In general, “fear”‐based disorders are often characterized by heightened physiological reactivity to salient threat stimuli, as measured by skin conductance response, fear‐potentiated startle, pupillometry, cortisol, alpha amylase, or heart rate variability. In contrast, “anxiety”‐related disorders are often characterized by a more blunted pattern of physiological reactivity using these same assays 202 . This is a potentially noteworthy dissociation providing some support to the notion that these biological measurements may be at least partly disorder‐specific and developed as potential biomarkers.
An exemplary longitudinal study demonstrated the utility of the error‐related negativity (ERN) as a specific, albeit not highly sensitive, electrophysiological biomarker predicting the first onset of GAD over 1.5 years in adolescent girls: ΔERN increased the odds of GAD onset to 1.64 even after controlling for clinical risk factors 203 . There is also evidence for altered interoceptive sensitivity as a marker or mechanism of anxiety disorders 204 : for example, hypersensitivity to carbon dioxide (CO2) introduced during a laboratory challenge has been proposed as a relatively specific and heritable predictive marker for subsequent panic attacks, but not necessarily panic disorder, during long‐term follow‐up 205 , 206 .
There are also pharmacological challenges that have the potential to serve as risk or diagnostic biomarkers, particularly in panic disorder. In one study, a yohimbine challenge increased panic symptoms in patients with panic disorder more than healthy controls, and the extent of the yohimbine‐induced symptoms in patients (but not controls) correlated with a trait measure assessing fear of publicly observable anxiety 207 . Similarly, another study showed that m‐chlorophenyl‐piperazine (mCPP) provoked panic symptoms in patients with panic disorder but not in those with generalized social anxiety disorder 208 , showing some diagnostic specificity. Finally, cholecystokinin tetrapeptide (CCK‐4) induces panic symptoms in individuals with panic disorder at a greater frequency than in healthy controls 209 , and the extent of symptom expression seems to be dose‐dependent 210 .
Predictive biomarkers of therapeutic response
Genetics
The 5‐HTTLPR/rs25531 variant in the SLC6A4 gene has been extensively explored in the context of psychotherapy‐genetic studies of anxiety disorders, which however have produced mixed results. A recent meta‐analysis incorporating 10 independent samples totaling 2,195 patients could not confirm a role of this genetic variant in moderating the effect of CBT on anxiety disorder outcomes 211 . Similarly, limited studies investigating other serotonin‐related (e.g., HTR1A, HTR2A, MAOA, TPH, TPH2), dopamine/noradrenaline‐related (e.g., COMT, DRD2, DAT1), or neurotrophic factor‐related (BDNF, NGF) genes did not report consistently replicable results 212 . The largest therapy‐genetic GWAS meta‐analysis of CBT treatment response in adults with anxiety disorders or major depressive disorder and in children with anxiety disorders (total N=2,724) failed to detect any sufficiently robust association of genetic variation with treatment outcome 213 . Finally, some recent studies have focused on potential epigenetic predictors of psychotherapy response in anxiety disorders, mostly pointing to a potential role of predictive DNA methylation patterns in the MAOA, SLC6A4, and FKBP5 genes 192 .
Pharmacogenetic studies in anxiety disorders have investigated candidate genes involved in pharmacokinetics (i.e., drug availability, metabolism and degradation) such as CYP2D6 and CYP2C19, or candidate genes involved in pharmacodynamics (i.e., receptors, transporters) of the serotonin, dopamine and noradrenaline systems; hypothalamic‐pituitary‐adrenal axis (HPA); stress pathways; and neurotrophic factors. Thus far, the results have been inconclusive 186 , 214 . One study used a commercially available test (NeuroIDgenetix®) in GAD 215 , but industry‐sponsored pharmacogenetic tests have yet to be implemented in daily clinical practice. To the best of our knowledge, only one GWAS to date has investigated genetic markers of treatment response to venlafaxine in GAD, but there was no genome‐wide significant association 216 . Pharmaco‐epigenetic research in anxiety is still in its infancy and has yet to yield any consistently promising findings 193 .
Neuroimaging
A systematic review of 17 studies on neuroimaging markers predicting psychotherapy response in anxiety disorders revealed the most compelling evidence for: a) mostly increased pre‐treatment dorsal anterior cingulate cortex activity during relevant fMRI tasks (e.g., emotional face processing/matching, anticipation of emotional pictures, or differential fear conditioning); and b) increased resting‐state anterior cingulate cortex‐amygdala coupling 212 .
Similarly, a quantitative meta‐analysis of primarily emotion processing/regulation task‐based fMRI studies observed that increased dorsal anterior cingulate cortex activity was related to CBT response in 17 datasets comprising 442 patients with various anxiety and stress disorders 217 . This meta‐analysis further revealed associations between treatment response and activations spanning the larger salience and interoception networks (i.e., comprising not only the dorsal anterior cingulate cortex, but also the right inferior frontal gyrus, anterior insular cortex, and dorsomedial prefrontal cortex). A sub‐analysis restricted to patients with social anxiety disorder revealed positive correlations between CBT response and activity of the bilateral Rolandic operculum, subgenual anterior cingulate cortex, right precentral gyrus, right dorsolateral prefrontal cortex, right supplementary motor area, and posterior cingulate cortex 217 .
Perhaps most promising of all, the first multimodal study integrating clinical data with resting‐state and structural brain connectomics imaging data using a machine learning approach predicted CBT outcome at a single‐subject level in social anxiety disorder with an accuracy of 84%; there was a five‐fold improvement in predictive power compared to clinical measures of severity and single connectomic measures alone 218 .
Additional studies have examined potential neuroimaging biomarkers of response to pharmacotherapy. An exemplary fMRI study in social anxiety disorder applying the Multi‐Source Interference Task revealed that greater pre‐treatment dorsal anterior cingulate cortex reactivity predicted better response to combined psychotherapy and selective serotonin reuptake inhibitor (SSRI) treatment with 83% accuracy 219 . In a pilot study of patients with the same diagnosis, higher baseline activity in the anterior and lateral parts of the left temporal cortex and the lateral part of the left middle frontal regions, measured by Tc‐99m HMPAO SPECT, predicted non‐response after a six‐eight week regimen of citalopram 220 .
Also in social anxiety disorder, a PET study discerned task‐based negative left amygdala/rostral anterior cingulate cortex and positive left amygdala/dorsomedial prefrontal cortex co‐activation patterns in SSRI responders. However, this study and further comparable investigations in other anxiety disorder phenotypes applied a longitudinal pre‐post design and thus followed a mechanistic rather than a predictive study approachsee 221 .
In GAD, a favorable response to eight‐week treatment with venlafaxine was predicted by greater rostral anterior cingulate cortex activity and lower amygdala activity in response to fearful faces, and by increased pregenual anterior cingulate cortex activity in anticipation of aversive and neutral images 222 , 223 .
Other measures
Cardiovascular markers – including tonic and/or phasic heart rate, heart rate variability, and blood pressure – as well as markers of the adrenergic system – including adrenoceptor density and plasma 3‐methoxy‐4‐hydroxyphenylglycol (MHPG) levels – have been proposed as potential markers of CBT response. Studies testing these biological assays, however, have largely used suboptimal study designs and/or relied on limited sample sizes 212 . Their potential to serve as biomarkers for anxiety disorders, therefore, remains to be determined.
Summary of anxiety disorder biomarkers
The majority of studies to date have failed to identify valid biomarkers of anxiety disorder pathogenesis or treatment response. Only a few studies have actually assessed sensitivity, specificity, or positive/negative predictive values 203 , 218 , 219 , 224 , 225 , and are potentially actionable. For instance, ERN showed a modest overall accuracy in predicting future GAD, with an AUC of 0.60, but marked elevations of ERN were quite informative about risk, and predicted onset above and beyond clinical risk factors 203 . Notably, because EEG can be widely deployed in clinical practice and the ERN marker is malleable by attention bias modification training 226 , this could be a promising and practicable risk marker of GAD.
Some task‐based MRI findings have also shown satisfactory prediction of treatment response 218 , 219 , 224 , 225 . Biomarkers based on connectomics neuroimaging methods (resting‐state fMRI, diffusion MRI) 218 might offer some additional advantages over activation mapping markers, as connectomics‐based measurements can be acquired more consistently and reliably across settings, are independent of task performance confounders, and can be performed even in infants 218 . It should be noted, however, that MRI‐based biomarkers ultimately might prove less clinically viable than neurophysiology‐based markers, since fMRI technology and analysis expertise are currently expensive and typically only available in large academic medical centers.
Apart from the few exceptions illustrated above, the majority of studies reported findings on a merely correlational or associative level, and did not assess sensitivity, specificity or positive/negative predictive values. According to the criteria for biomarker discovery 11 , markers of anxiety disorder pathogenesis and treatment proposed so far remain at stage 1 (“target identification”) and have not explicitly ruled out confounding factors such as stress, comorbidity, physical activity and/or psychotropic medication (stage 2, “internal validation”). Most presently available findings also warrant replication in validation samples independent of the discovery samples (stage 3, “external validation”), and await testing in trials demonstrating “clinical utility” (stage 4). Therefore, their potential remains unclear 223 .
BIOMARKERS IN POST‐TRAUMATIC STRESS DISORDER
A distinct literature has developed on biomarkers for PTSD. This research frequently operationalizes post‐traumatic stress with continuous scores on measures such as the PTSD Checklist (PCL‐5) 227 . The most well‐established pharmacotherapies for PTSD are SSRIs, but they have limited efficacy 228 . Consequently, PTSD research has focused on developing susceptibility and diagnostic biomarkers.
Genetics
GWAS have revealed that contributions of individual genetic polymorphisms to PTSD are very small 229 . For example, in the largest GWAS to date (48,221 individuals with PTSD and 217,223 without), the effect size of the top single‐nucleotide polymorphism (SNP) was trivial (odds ratio, OR=1.06) 230 . However, PRS that combine effects of hundreds of thousands of SNPs show meaningful, albeit modest effects. The strongest PRS was correlated at r=.20 with the PTSD Checklist for DSM‐5 (PCL‐5) score. This estimate is likely optimistic, because it was assessed using internal replication. Other studies have observed effect sizes of r=.10 to r=.16 in independent samples using an earlier version of this PRS 231 , 232 .
Neuroimaging
Numerous neuroimaging studies have investigated potential biomarkers of PTSD, but they have produced mixed results, also due to small sample sizes 233 . Meta‐analyses have revealed clear links of PTSD to smaller hippocampal, amygdala and total brain volume, and lower structural connectivity of the corpus callosum 234 , 235 . Few studies examined other regions, making conclusions about them less reliable. Moreover, meta‐analyses are vulnerable to publication bias (e.g., primary studies reporting only on regions where significant effects were found).
Mega‐analyses can address this limitation by pooling voxel‐level data from multiple samples to perform whole‐brain analyses and present an unbiased picture of neural correlates. The Enhancing NeuroImaging Genetics through Meta‐Analysis (ENIGMA) consortium performed a mega‐analysis of cortical volume comparing 1,379 people with PTSD to 2,192 without 236 . Significant differences were found in 21 regions, all showing smaller volume in PTSD, but the largest group difference was only 0.17 standard deviations (SD). Another ENIGMA study examined subcortical volumes in 794 individuals with PTSD and 1,074 without 237 . Hippocampal volume was lower in PTSD by 0.17 SD. Amygdala volume and total brain volume were also smaller, but these differences did not reach significance. ENIGMA mega‐analysis was also conducted on structural connectivity in 1,426 people with PTSD and 1,621 without 238 . Significant differences emerged in only one region, tapetum of corpus callosum, where structural connectivity was lower in PTSD by 0.11 SD.
Overall, these ENIGMA studies used rigorous methodology, and alignment with previous meta‐analyses further increases confidence. However, their results have not been confirmed in sufficiently powered independent studies yet, and their relevance for developing biomarkers of susceptibility, prediction, or treatment response is questionable.
As links between PTSD and regional structural morphology are weak, at least for standard neuroimaging modalities, one potential avenue for improvement is if regions or voxels are combined into a composite biosignature, especially using machine learning techniques. Reliable composites will likely require larger sample sizes than currently available for PTSD. For example, research to estimate a participant's age from sMRI found that a biosignature developed in a sample of 35,474 individuals correlated at r=.69 with age in an independent sample 239 . Performance of this biosignature was proportional to the sample size used to develop it, although most of the increase occurred by the sample size of 5,000.
Peripheral biomarkers
Numerous peripheral measures have been explored as potential biomarkers of PTSD, and we consider only the most studied ones.
Psychophysiology research has linked PTSD to autonomic nervous system functioning. Meta‐analyses of heart rate variability and respiratory sinus arrhythmia found differences between people with and without PTSD of 0.16 to 0.50 SD 240 , 241 , though some of this signal may be due to potential confounders such as low socioeconomic status or physical comorbidities. Another possible marker is startle response to an acoustic probe (i.e., sudden loud sound) when the person expects danger (e.g., possible electric shock) (fear‐potentiated startle). This paradigm produces reliable differences in startle magnitude between danger and safety conditions, but the link to PTSD is unclear. It appears that some patients with PTSD show potentiation, while others are indistinguishable from healthy participants 242 . This casts doubt on fear‐potentiated startle as a viable biomarker.
Biomarker research has also focused on developing relevant assays from blood, urine and saliva. Evidence suggests that these assays are informative, albeit imperfect, surrogates for brain tissue 243 , 244 , 245 , 246 . Initially, peripheral tissue studies focused on candidate markers, such as cortisol and inflammation markers 247 , 248 . They reported elevated inflammation in PTSD, but the findings were quite mixed and may have been confounded by physical comorbidities.
Omics studies seek to investigate tissues comprehensively. The largest methylome‐wide study included 878 participants with PTSD and 1,018 without, finding four significant methylation sites, all in gene AHRR 249 . Two of these sites were replicated in an independent sample 250 . However, the difference between groups was small even for the top site, and a composite methylation signature was not attempted.
The largest transcriptome‐wide study included data on 977 participants and did not find any significant associations between PTSD and expression of individual genes 251 . However, it did not attempt to develop a transcriptomic signature. Two previous studies constructed such signatures and observed moderate accuracy, with an AUC of 0.64 to 0.76 252 , 253 , but neither signature was evaluated in an independent sample.
Proteomics is a new modality in PTSD research. The most comprehensive study to‐date included 276 plasma proteins, finding significant links between numerous proteins and PTSD 254 . A multiprotein signature showed a moderate association with PTSD within‐sample, but these results require replication.
Summary of PTSD biomarkers
PRS for PTSD has passed the first three stages of biomarker development. It is an informative measure of susceptibility, reliable and non‐invasive. However, its links with PTSD are too weak to be useful clinically. Even as performance of PRS improves with increasing discovery sample sizes, it is unlikely to reach the level needed for clinical utility. Nevertheless, it may become useful when combined with clinical and demographic risk factors 255 .
The other potential biomarkers need rigorous replication and careful control for confounds. Moreover, their links to PTSD are quite weak. Integration of data across the whole brain, methylome, transcriptome and proteome is needed to improve effect sizes and replicability. However, this approach requires very large sample sizes.
BIOMARKERS IN MOOD DISORDERS
Mood disorders (MDD and BD) are among the most prevalent and costly illnesses 167 , and the development of clinically actionable biomarkers is therefore a critical research and therapeutic goal. Early biomarker efforts throughout the 1970s and 1980s centered on urinary levels of various catecholamine metabolites, measures of platelet monoamine oxidase activity, and hypothalamic‐pituitary‐adrenal axis (HPA) function 256 , 257 , with inconclusive results.
Neuroimaging modalities, such as gaseous encephalography, were used as early as the 1950s 258 , and structural imaging methods were increasingly employed throughout the 1980s in attempts to define major structural and volumetric differences in the brains of individuals suffering from mood disorders 259 . The use of neuroimaging in mood disorders dramatically increased in popularity over the last 20 years, with the introduction of novel, minimally invasive MRI and MRS techniques that allow for structural, functional and neurochemical investigations. These emerging modalities were paired with novel pathophysiological and treatment concepts such as neuroinflammatory pathology, neurometabolic contributions to behavioral disorders, circuit/network based disorders, and neuroplasticity enhancing treatments. The field of psychiatric genetics has also leveraged technical and conceptual advances to perform ever larger studies aiming to identify genetic variation contributing to disease risk and treatment response.
However, despite decades of work attempting to identify clinically meaningful diagnostic tools and procedures, there has been limited progress in developing biomarkers that meaningfully aid in the diagnosis, prognosis, or personalization of treatment choices for mood disorders. Below, we present an overview of the current state of knowledge.
Susceptibility biomarkers
Genetics
The hope of identifying genetic biomarkers for mood disorders is bolstered by the clear epidemiological evidence of heritable factors contributing to these disorders, such as the moderate level of genetic contribution found in the Scottish Family Health Study for MDD 260 , and the heritability estimates of approximately 70‐90% for BD 261 . However, the identification of specific genetic contributions to these disorders has proven extremely challenging.
Several large, recently‐completed GWAS have provided new information on the genetics of mood disorders. For example, a GWAS study identified 17 loci – relating to aspects of brain function ranging from excitatory neurotransmission to neuron spine/dendrite functions – that were associated with depressive phenotypes in ~114,000 people 262 . These findings are generally in line with a meta‐analysis of post‐mortem studies, which reported lower levels of synaptic protein or mRNA across MDD and BD (protein levels of SNAP‐25, PSD‐95 and syntaxin in MDD, and PSD‐95 mRNA levels in BD) 263 .
More recent GWAS efforts with growing sample sizes continue to expand the number of genome‐wide significant hits 264 , 265 . These large studies provide additional evidence that the genetics of depression maps onto the broader genetic structure of mental disorders and cognition. Furthermore, transcriptional signatures in MDD have been found to be gender‐specific 266 , 267 , suggesting an important further area of investigation.
A GWAS study in ~42,000 patients identified 15 genes linked to BD 268 . However, the pattern that emerges most consistently is that BD shares many common weak genetic risk factors with MDD and schizophrenia 269 . Ultimately, it seems likely that these genes will each have a small impact on the susceptibility to mood disorders, and progression to disorder will importantly depend on interactions with environmental factors, such as (perceived) uncontrollable stress.
Neuroimaging
Much of the neuroimaging work related to mood disorder susceptibility seems to parallel the effects of stress on the central nervous system. Large meta‐analyses of the structural correlates of mood disorders show volume reductions in the areas commonly associated with the stress response, including the hippocampus and frontal lobe 259 .
The large multi‐site ENIGMA consortium study, comprising ~2,000 participants, found reduced hippocampal volume, lower cortical thickness in multiple regions, and white matter alterations in MDD 270 . Similar to the genetic studies, the ENIGMA findings indicate relatively small overall effect sizes and common abnormalities in MDD, BD and schizophrenia, making it unlikely that the measures will prove useful in providing diagnostic biomarkers at the individual level. However, newer work using PET and MRI methodologies may be uncovering the cytoarchitectural correlates of the above structural findings, by demonstrating reductions of synaptic density in MDD 271 .
Functional imaging studies, conducted at rest or during the performance of emotional or cognitive tasks, have increased dramatically over the last decade. Although these studies have provided support for circuit and network contributions to mood disorder pathophysiology, few findings reliably distinguish mood disorders from other conditions 272 , similar to the structural and genetic findings previously described. Overall, prefrontal dysfunction may be a trait feature of mood disorders, while increased activation in limbic regions such as the anterior cingulate cortex and the amygdala may be associated with symptom expression 273 . However, medication and the presence of comorbid conditions – such as anxiety or substance abuse – may affect these findings. Much more work is needed at both the technical and conceptual levels before functional imaging can have more direct clinical applications in mood disorders.
The findings of MRS studies are far from consistent or conclusive, and are often complicated by methodological and technical heterogeneity. However, they have provided evidence to suggest the involvement of the amino acid neurotransmitter systems, including GABA and glutamate, in the pathophysiology of mood disorders 274 , 275 , 276 , 277 . These findings again appear not to be pathognomonic of these disorders, but common among a range of psychiatric conditions 278 , 279 , 280 , 281 , 282 .
Early‐stage MRS studies also suggest a role for pathophysiological effects related to oxidative stress as a contributor to mood disorders. Newer proton‐MRS has enabled the quantification of glutathione concentrations in the brain, allowing for in vivo investigations of oxidative stress neurochemistry 283 . Although only a limited number of MRS studies have been completed to date, there is some indication of reduced glutathione measures in mood‐disordered patients compared with healthy controls, with possible differences between MDD and BD 284 , and specific regional associations with anhedonia in MDD 285 . Most recently, glutathione concentrations in the anterior cingulate cortex were found to be inversely correlated with depression scores and white matter hyperintensities associated to COVID‐19 infection, further suggesting a link between neuroinflammation, oxidative stress, and mood 286 .
In sum, the emergence of novel MRS methodologies has allowed unique in vivo explorations related to neurochemistry, brain metabolism, and neuroenergetics of mood disorders. While various technical and methodological limitations of the existing studies prevent firm conclusions about relationships to mood disorder susceptibility, the emerging data are generally in line with rodent models and post‐mortem studies 287 , 288 , and with predictions from large genetic studies showing effects on synaptic density and excitatory/inhibitory neurotransmitter pathways 265 , 268 , and may reflect the reduction of synaptic density observed in MDD 271 . However, the methodology to date is not meaningfully helpful in providing prognostic markers, or in differentiating clinically relevant phenotypes of mood disorders.
Peripheral biomarkers
A myriad of peripheral measures attempting to capture monoaminergic neurotransmitter functioning have been employed over the years, with limited to no success in modifying diagnostic or treatment approaches. More recently developed metabolomics approaches, however, allow dynamic measures of large numbers of metabolites over time. Meta‐analyses of these studies have found several metabolic abnormalities associated with mood disorders, including decreased levels of tryptophan, kynurenic acid and kynurenine, and increased glutamate levels in MDD patients. Pathway and network analyses of these data indicate disturbances of amino acid and lipid metabolism, especially the tryptophan‐kynurenine pathway and fatty acid metabolism 289 . Overall, these findings support the involvement of several pathophysiological processes – including cellular signaling systems, components of the cell membrane, various neurotransmitter systems, hormonal regulation, moderators of circadian rhythm and sleep, as well as inflammation and immunological factors. However, no specific abnormalities can be associated with clinically meaningful differences to date.
Peripheral HPA axis measures have also been studied in large numbers of patients over the last five decades. The findings generally indicate HPA hyperactivity in depression, providing evidence of a link between MDD and comorbid conditions such as diabetes, dementia and coronary heart disease, especially in older and more severely depressed inpatients with melancholic or psychotic features 290 . However, the results are quite heterogeneous, and the effect sizes are modest, limiting the utility of these measures as diagnostic tools 291 .
A relatively large number of studies, conducted over the past two decades, have examined peripheral measures of relevant neurotrophic factors. Much of this work has demonstrated a relationship between abnormally low peripheral measures of brain‐derived neurotrophic factor (BDNF) and mood disorders. Reduced plasma and serum BDNF levels are commonly reported in depressed patients and may be altered with treatment response 292 , 293 . Although fitting nicely with data suggesting that BDNF gene expression and function contributes to the pathophysiology of depressive‐like behaviors, these findings lack specificity, and notable inconsistencies pervade research related to gene expression changes in human and rodent studies 294 .
Interest in peripheral markers of immune function and inflammation has also increased over time. Abnormalities in several inflammation markers – including C‐ reactive protein (CRP), IL‐6, IL‐12 and tumor necrosis factor alpha (TNFα) – are consistently observed in major depression, often with medium sized effects. Nevertheless, the specificity and selectivity of these markers has not yet been convincingly demonstrated 295 .
Finally, an emerging but potentially exciting body of research has begun to examine susceptibility biomarkers related to circadian rhythms. Mounting evidence suggests that abnormalities in circadian phase may precede mood disorders 296 , 297 , 298 , 299 , 300 , 301 , 302 , 303 , 304 . Indeed, change in sleep is one of the diagnostic criteria for MDD 305 , and sleep difficulties are present in >85% of cases 298 , 306 , 307 , 308 and are correlated with greater depression severity 309 , 310 . Depressed patients exhibit reduced motor activity during the day 309 , 311 , and lower rhythm‐adjusted mean (or midline estimating statistic of rhythm, MESOR) has been reported as a diagnostic indicator of depression, with up to 80% accuracy 312 . A phase shift in activity is also present, as time to peak activity (acrophase) is delayed 309 , 312 , 313 , and this delay may be associated with greater depression severity 309 , 313 . Disrupted sleep is also a diagnostic criterion for BD 305 , 314 . BD patients are also likely to have lower amplitude of motor activity when depressed 315 or euthymic 316 .
Interestingly, circadian markers may have the potential to distinguish MDD from BD 317 . In particular, while further studies are needed, initial evidence suggests that MDD is associated with a phase delay 318 , 319 , whereas BD is associated with a phase advance during mania 320 , and a phase delay during euthymia 321 , mixed mania, and depression 320 , 322 . Additional information may be gleaned by measuring melatonin during sleep/wake cycles 304 , 323 , 324 , or by measuring awakening cortisol levels in euthymic BD patients 325 .
Further work has indicated that the 3111T/C clock gene polymorphism, related to circadian rhythms, is associated with a higher recurrence of initial, middle and early insomnia in homozygotes for the C variant and a similar trend concerning decreased need of sleep in BD patients 326 . Moreover, expression of the clock genes PER1 and NR1D1 from saliva of BD patients in manic episodes is phase advanced relative to depressive episodes 327 .
Prediction of treatment response
The best predictors of treatment response in mood disorders have traditionally included demographic and clinically‐defined factors, such as age, severity and duration of illness, number of comorbidities, and the presence or absence of psychotic or mixed features 328 , 329 . However, several large‐scale efforts aiming to identify biological predictors of treatment response have been implemented in the last decade. The international Study to Predict Optimized Treatment of Depression (iSPOT‐D) 330 , the Predictors of Remission in Depression to Individual and Combined Treatments (PReDICT) 331 , and the Genome‐Based Therapeutic Drugs for Depression (GENDEP) 332 studies all used large sample sizes to study and identify possible biomarkers for treatment response in depression.
The iSPOT‐D study randomized over 1,000 depressed patients to 8 weeks of treatment with escitalopram, sertraline or venlafaxine, and assessed a large number of outcome variables, including clinical, daily functioning, cognitive, genetic and psychophysiological (e.g., EEG, event‐related potentials, heart rate) markers. At least some outcome predictive value was found for various EEG measures (such as alpha connectivity) 333 ; genetic factors (including variants in the CRHBP gene 334 , and regulators of the gene coding for P‐glycoprotein, which limits brain concentrations of certain antidepressants 335 ); structural markers (such as hippocampal tail volume 336 ); functional measures (such as activation in the frontoparietal network during response inhibition 337 ); and environmental factors (such as early childhood trauma 338 ). Further evidence also suggested that measures of functional connectivity of cognitive control and reward circuits could selectively and differentially predict antidepressant treatment responses 339 , 340 .
The GENDEP study identified baseline levels of macrophage migration inhibitory factor (MIF), IL‐1β, and TNF‐α as “predictors” of antidepressant treatment response. That is, higher levels of pro‐inflammatory cytokines predicted lack of antidepressant response, but lower levels did not predict a positive antidepressant response 341 . Interestingly, modulation of the glucocorticoid receptor complex and measures related to neuroplasticity were associated with a therapeutic antidepressant effect.
The Establishing Moderators and Biosignatures of Antidepressant Response for Clinical Care for Depression (EMBARC) study was a randomized sequential treatment study. In phase 1 of the study, nearly 300 patients were randomized to either sertraline or placebo. In phase 2, non‐responders were randomized to placebo, sertraline (except previous non‐responders), or bupropion. The goal was to identify potentially treatment‐predictive endophenotypes of MDD, using electrophysiological methods, functional imaging, and peripheral markers 342 , 343 , 344 . Pre‐treatment brain reward was somewhat predictive of individual antidepressant response 345 . Similar to reports from the CO‐MED trial 346 , the study also found evidence suggesting that CRP levels had some value in predicting treatment response, though the effect may be gender‐specific 347 .
The familial aggregation of BD (particularly the lithium‐responsive type) among responders to lithium prophylaxis 348 has prompted recent attempts to identify genomic correlates of lithium response. The largest GWAS study 349 found a single associated locus on chromosome 21. The authors demonstrated that the response‐related alleles were associated with lower rates of relapse in an independent sample of 73 patients treated for two years of lithium monotherapy. However, the predictive power of genomic data remains unknown, since GWAS are not designed to evaluate predictive capacity 350 .
Summary of mood disorder biomarkers
Numerous biomarkers have been proposed in MDD and BD, and multiple studies including large‐scale initiatives have identified potential candidates for future validation. There are several reasons underlying the limited gains achieved to date. These include the extreme heterogeneity of mood disorder diagnoses, limited treatment options, incomplete understanding of how the biomarkers actually reflect pathophysiological states, and the specific technical limitations and costs of the individual modalities.
Neuroimaging modalities, particularly those based on emerging spectroscopy approaches, may hold promise for improving prediction. However, at present, it remains a challenge to distinguish such findings in MDD or BD from similar effects in other mental disorders, such as schizophrenia, perhaps partly due to the imaging measures capturing a generalized “stress” phenotype rather a disorder‐specific signature.
Similar challenges of sensitivity and specificity confront the use of peripheral biomarkers, such as neurotrophic factors as well as markers of immune function and inflammation. Future research may aim to integrate these measures with emerging approaches, such as those related to circadian rhythms, which may have the capability of differentiating MDD and BD biologically and behaviorally.
Acquiring combinations of available markers may also be useful 351 , 352 , 353 , potentially providing complementary information while reducing noise and alternative explanations that compromise internal validation efforts. Future studies will also need to continue unraveling the formidable heterogeneity in mood disorder prediction, which may depend on factors such as gender and early childhood adversity, among many others.
BIOMARKERS IN SUBSTANCE USE DISORDERS
Among psychiatric disorders, SUDs are unique in the sense that the presence of a potentially addictive drug within the context of use‐related dysfunction provides biological evidence for a disorder. Yet, beyond the presence of a drug in individuals’ biological fluids, SUDs reflect repeated, compulsive substance use patterns mediated by complex (and not well‐understood) interactions between biological drug effects, genetic predispositions, early life experiences, external stressors, and central and peripheral nervous system adaptations. Only an estimated 1 in 7 individuals who use substances will progress to use disorder 354 , and such estimates vary according to which substance is used 355 . Considering that stable use with limited adverse consequences may describe a substantial group of substance‐using individuals 356 , it is vital to determine which susceptible individuals will eventually transition into full SUD.
Furthermore, among those diagnosed with SUD, there is a huge treatment gap. Epidemiological data indicate that 19 out of 20 individuals classified as needing treatment for SUD do not think that they need it 357 , and this low perception of treatment need tends to persist over time 358 . Multiple factors likely drive this low perception, including minimization of the signs/symptoms of dysfunction associated with substance use, fear of stigmatization 359 , and possibly neurocognitive impairments that might affect insight 360 , 361 . Thus, it is vital to develop and validate biomarkers that can help predict who is more likely to engage and succeed with treatment.
The area of neuroimaging has attracted the greatest attention in the search for biomarkers in SUDs 362 , 363 , for several reasons. First, there are now well‐established substance‐related brain circuitry changes that involve: a) disruption in executive control, underpinned by dysregulation of prefrontal‐subcortical processing, and b) increased salience processing of substance‐related stimuli, spanning subcortical and cortical structures 364 . Second, neuroimaging can be used to integrate mechanistic models of substance effects (e.g., increasing dopaminergic tone via the transporter 365 or vesicular sources of intracellular dopamine 366 ) with adaptations of brain systems to repeated use (e.g., decreased dopamine receptor availability and release measured with PET), which are some of the most reliable findings in the addiction literature 367 , 368 . Third, some studies support the idea that individual differences in neuroimaging assays may be of sufficient effect size to be used for predictive purposes 369 . On the other hand, several studies show clear evidence of limited associations between brain and behavior 370 , and even large‐scale studies show only modest correlations between psychopathology and structural or functional brain characteristics 371 . Despite these challenges and uncertainties, several studies have successfully used neuroimaging markers to predict important substance use outcomes.
Susceptibility biomarkers of transition to problematic use
In one large cohort study, transition from no use to frequent drinking in early to mid‐adolescence was predicted by blunted activity of the medial orbitofrontal cortex during reward outcome 372 . Also in adolescents, structural characteristics – such as a larger cingulate gyrus – were predictive of resilience to problematic use after 3 years 373 . In a review of 44 longitudinal neurobehavioral studies predicting substance use in youth 374 , functional vulnerability markers of substance use – i.e., markers that predict onset of subsequent substance use – included increased fMRI activation during reward feedback and risk evaluation in prefrontal and ventral striatal regions, and fronto‐parietal hypoactivation during working memory. Altered neural patterns during response inhibition and differences in structural markers, including smaller fronto‐parietal and amygdala volumes and larger ventral striatal volumes, were also observed.
In one examplary study of this approach, occasional stimulant users completed a Risky Gains Task during fMRI and were followed up to three years later to determine whether or not they transitioned to problematic use. Compared with participants who stopped their occasional stimulant use, those who later transitioned to problematic use made riskier baseline decisions after winning feedback, and exhibited lower baseline frontal, insular and striatal blood oxygen level dependent (BOLD) responses to win/loss feedback after making risky decisions 375 . In another study, initially alcohol‐naïve adolescents were tested with fMRI during a reward task and were followed for 3 years to determine whether or not they initiated alcohol drinking. Compared with adolescents who did not initiate alcohol use, those who did displayed increased baseline fMRI activation to loss in the left dorsal striatum (putamen) and right precuneus 376 . Finally, a study used connectome‐based predictive modeling with leave‐one‐out cross‐validation to uncover stress‐linked connectivity patterns that differentiated risky from non‐risky drinkers, finding that the stress‐linked network profiles of the risky drinkers predicted loss of control of drinking in the entire sample 377 .
These brain‐based assays complement well‐established behavioral predictors of problematic substance use. Investigators have found that non‐planning and affect‐based impulsivity, as well as reward‐related valuation, were predictors of SUD vulnerability 378 . Other investigators have additionally found support for relatively poor control, early substance use initiation, binge patterns of use 379 , and lower efficiency of evidence accumulation 380 . However, regardless of whether these neuroimaging or behavioral markers can serve as predictive biomarkers, a fundamental question persists of how these markers can be used in a predictive context. In fact, no study in SUD has used prospectively predictive biomarkers to examine their ability to affect outcomes or guide clinical decisions.
Predictive biomarkers of relapse and abstinence
Among individuals who have already met criteria for SUD, another important goal for biomarker development is the prediction of relapse versus abstinence. Prediction of relapse to substance use has been a major focus for biomarker development in SUD research for some time 381 , and several investigators have been intrigued by findings that individual differences on various biological or behavioral measures provide predictive information 382 , 383 , although there is increasing recognition that such outcomes are only a small part of the many possible clinically relevant measures 384 , and that a comprehensive approach using complementary outcomes for prediction has been missing.
In a cue‐reactivity study conducted among individuals with alcohol use disorder, fMRI activation in the ventral striatum during viewing of alcohol vs. neutral pictures predicted a shorter time to relapse 385 , perhaps reflecting a heightened vulnerability to drug cues that culminates in drug‐seeking and drug‐taking behavior during abstinence. Consistent with this, an EEG study was conducted in a cohort of individuals with cocaine use disorder who were subsequently subgrouped based on length of cocaine abstinence. Results showed that the EEG‐measured late positive potential (LPP), acquired in response to viewing cocaine images (and previously linked to craving 386 ), showed a quadratic (inverted U‐ shaped) pattern as a function of abstinence length 387 . These results were interpreted as potentially reflecting an objective biological marker of craving “incubation”, where drug‐ cue reactivity is counterintuitively potentiated after short‐ and medium‐term abstinence 388 , 389 .
In cue‐reactivity research, one potentially important consideration is the contrast examined. While many studies have historically tested a standard drug vs. a neutral contrast, a drug vs. a pleasant contrast may be more ecologically valid 390 , 391 . This latter contrast better reflects diagnostic criteria, for example as specified in the DSM‐5, where time, effort and resources need to be allocated toward the pursuit and consumption of the addictive drug at the exclusion of other activities 392 , 393 . They also better tap into theories of addiction that emphasize a shift in salience and hedonic value from pleasant reinforcement to drug reinforcement 394 , 395 .
Beyond cue‐reactivity, other studies have reported that reduced functional resting‐state connectivity within the executive control network, and between the executive and salience networks, could serve as a marker of relapse risk 396 . There may also be important clinical outcome predictive effects of brain areas involved in inhibitory control, such as the inferior frontal gyrus, dorsal anterior cingulate cortex, and dorsolateral prefrontal cortex 397 , as well as brain areas involved in stress reactivity, such as the ventromedial prefrontal cortex and ventral striatum 398 . For stress reactivity, a recent study of patients with alcohol use disorder first found case‐control differences (i.e., ventromedial prefrontal cortex and ventral striatal hypoactivity to stressful, threatening images) (Study 1), and then found that these same imaging phenotypes predicted a faster relapse to drinking in a new sample (Study 2) 399 . The finding of clinical implications in a new sample lends support to this imaging phenotype as a target for biomarker development into the future.
An alternate, but related strategy has been to examine functional and structural correlates of sustained abstinence 400 . A recent systematic review concluded that drug abstinence tracked with increased gray matter volume in multiple cortical regions spanning the frontal, temporal, parietal and occipital lobes. There were also volumetric increases in the insula, cerebellum, hippocampus and thalamus, though not the striatum 400 .
Functional studies were relatively less clear, though some evidence indicates changes in subcortical areas such as the midbrain and striatum. For example, an earlier study scanned individuals with cocaine use disorder using a drug‐Stroop task that incorporated monetary payouts for correct performance. Results showed that midbrain activation during this rewarded cognitive control task increased from baseline to six‐month follow‐up, during which participants were abstinent and/or treatment‐seeking. Furthermore, the more the midbrain task activation increased from baseline to follow‐up, the greater was the reduction in scores on a simulated drug‐seeking task 401 that itself has been associated with real‐world drug use/severity 402 , 403 and dopamine D2‐type receptor availability 404 .
The finding of midbrain activation enhancement to monetary reward in cocaine addiction was since independently replicated: relative to pre‐treatment measurements, post‐treatment was associated with increased activity in anticipation of reward in the midbrain, thalamus and precuneus; and increased activity in midbrain correlated with one‐year cocaine abstinence, while an increase in ventral striatal activity during loss anticipation correlated with fewer negative urine screens 405 . Insula functioning may also play a role, depending on the task: in one examplary study with methamphetamine‐addicted individuals in early abstinence, insula fMRI activation during risky decision‐making predicted relapse with greater sensitivity and specificity than standard clinical variables (e.g., days since last use) 406 . Studies like these highlight the utility of imaging phenotypes as complementary markers of relapse prediction, but further development and validation needs to occur before they may be deployed as clinically‐actionable biomarkers.
The application of rigorous machine learning approaches to avoid overfitting has provided evidence that it should be possible to use markers for disease stage prediction. In a recent review, a state‐of‐the‐art approach was proposed to develop predictive biomarkers within the context of relapse, which entailed computing connectivity patterns that generated out‐of‐sample predictions of outcomes 369 . For example, in one of the studies 383 , cocaine abstinence was predicted by increased connectivity between frontoparietal and medial frontal networks; increased connectivity among salience, motor/sensory and subcortical networks; and decreased connectivity between these two systems. Importantly, the results were replicated in an external sample, providing some predictive validity. Such studies and perspectives provide a blueprint for addiction biomarker research into the future.
Summary of substance use disorder biomarker research
The development of biomarkers for addiction has been impeded by multiple difficulties. First, as this field is still maturing, study designs have been largely limited to case‐control or longitudinal follow‐up approaches, and even most longitudinal studies predicting substance use outcomes have not provided additional data or methodologies confirming the predictive value of the measurements. Randomized intervention trials, such as neurostimulation approaches that directly modulate addiction‐related neural signatures 407 , will be important to use prospectively in order to examine the utility of these markers.
Second, addiction describes the compulsive use of different classes of substances that, while usually acting upon a largely common final neural substrate, have fundamental differences in their mechanisms of action and treatment approaches – even for different classes of “stimulants”, encompassing cocaine and methamphetamine 408 . This substance heterogeneity also complicates the search for genetic biomarkers in addiction. For example, as much as 38% of the variation in opioid addiction may be due to genetic factors specific to opioids (i.e., not shared with other substances) 409 , 410 .
Third, different biomarkers may be more appropriate depending on a person's current stage of addiction trajectory. For example, a large body of preclinical work has shown that initial drug‐taking may be largely mediated by ventral striatal (“reward”) pathways, whereas later‐stage addiction may be largely mediated by dorsal striatal (“habit”) pathways, and some human neuroimaging work has revealed a similar pattern of results 411 , 412 , 413 .
Fourth, arguably more than in other psychiatric disorders, environmental exposure is important, as someone can never become addicted if he/she never tries a particular substance. As an illustrative example, estimates have suggested that as much as 62% of the variance in cannabis misuse was shared with cannabis initiation 412 , 414 , highlighting the importance of substance availability for the eventual addiction phenotype. It should be noted that one exceptional counterexample is the alcohol dehydrogenase genes protecting against alcohol consumption and dependence 415 .
Fifth, SUDs do not exist in isolation, but frequently co‐occur with other behaviors and mental health conditions that are likely to have a profound influence on various biological markers. For example, most clinical studies of SUDs, by necessity, allow some amount of commonly occurring psychiatric comorbidity, such as depression or PTSD. Such constraints argue against the utility of potentially accessible, scalable and affordable – but largely non‐specific – biomarkers such as genetic or blood‐based measures, because these kinds of non‐specific assays may be difficult to interpret mechanistically.
In this vein, recent work suggests that extracellular vesicle‐associated miR‐29a‐3p plays a crucial role in methamphetamine use disorder and might be used as a potential blood‐based biomarker for detecting chronic inflammation and synaptic damage 416 . However, blood‐based measures indicating substance‐induced metabolic or inflammatory changes 417 may be additionally influenced by poor lifestyle (e.g., lack of exercise, poor diet and disrupted sleep) and other mental health conditions (e.g., comorbid mood or anxiety disorders), and so specificity remains a concern.
Nevertheless, an objective marker of substance use severity, not unlike a blood glucose or hemoglobin A1c measurement, might help to facilitate evaluation of treatment need (among clinicians) and treatment acceptance (among patients), and could be a low‐hanging fruit in the search for biomarkers in SUDs if an appropriate biomarker could be devised and confirmed for efficacy.
GENERAL DISCUSSION AND RECOMMENDATIONS
A review of candidate biomarkers for predicting diagnosis and treatment responsivity in ASD, schizophrenia, anxiety disorders and PTSD, MDD and BD, and SUDs, encompassing genetic, molecular, neuroimaging and/or peripheral phenotypes, reveals that most are in the very early stages of development and validation, and, for this reason, an assessment of their clinical utility is premature.
The immature state of the biomarker development in mental disorders is unsurprising, given its many challenges. One of the most fundamental challenges to overcome is that psychiatry research to date has most often relied on case‐control designs, contrasting behavioral and/or biological assays between patients with chronic illness and healthy comparison participants. This is even the case for large‐scale imaging datasets such as ENIGMA and similar consortia. Such consortia have been, and will likely continue to be, important for summarizing statistically robust group differences and associations in patients and controls, and will have the power to illuminate novel brain‐behavior findings that might otherwise remain hidden. Nevertheless, consortia data built from case‐control studies are unlikely to deliver clinically applicable biomarkers for relevant clinical indications understood in terms of the FDA, because the constituent case‐control studies themselves generally lack an actionable biomarker target (i.e., not diagnosis, but rather susceptibility or treatment response).
Our perspective is that studies should be designed from the outset to have as their designated end goal the development of a biomarker for a particular indication, in the right population for that indication (e.g., CHR for conversion, drug‐naive adolescents to predict development of SUD, acute unmedicated psychotic patients to predict response to first‐line treatments). In this way, psychiatry as a field can begin to move away from pursuing large studies relying on non‐specific big‐data approaches, and instead pivot toward designing a pipeline for a given target in a manner more akin to drug development. The Adolescent Brain and Cognitive Development (ABCD) study 418 has the potential to drive new biomarker knowledge on SUD susceptibility, for example. The PRONIA consortium has the potential to advance biomarker development for CHR for psychosis. Even then, it is not the case that massively large datasets are necessarily required for biomarker development; smaller studies could suffice, provided that a particular biomarker produces sufficiently high discrimination accuracy for its indication.
Another fundamental challenge for biomarker development in psychiatry is the heterogeneity of the disorders themselves. The diagnostic criteria for psychiatric conditions continue to rely on constellations of symptoms and signs that group patients together even if they exhibit very different illness presentations. For example, there are ~1,500 combinations of symptoms that result in a diagnosis of MDD 419 . Therefore, the cohorts of individuals we study are necessarily heterogeneous, and samples in which biomarkers are developed may not be representative of the larger population of patients who meet criteria for a given diagnosis. While newer diagnostic and classification systems, which are more quantitative and/or biologically‐based 420 , 421 , may help address this concern to some degree, a fundamental impediment to progress pertains to our insufficient knowledge of the human brain, and this likely can only be fully addressed with novel technologies that fundamentally increase the precision of measurement. Moreover, biomarkers will still be subject to potential confounds – including medication status, age and gender, among others – which threaten internal validity. For neuroimaging in particular, there are even more factors to consider, such as variability stemming from the processing pipeline 422 .
Despite these formidable challenges, several strategies and recommendations – either considered individually or in combination – may improve sensitivity and specificity in the search for biomarkers of pathogenesis, treatment response, and safety in psychiatric disorders. First, adequately powered collaborative (epi)genetic studies are needed, involving mega‐samples of patients who are deeply phenotyped for clinical course and treatment response, thereby allowing for the generation of poly(epi)genic risk in combination with poly‐environmental scores. PRS have been developed for many forms of psychopathology, but must continue to be updated and refined as more GWAS data become available; other psychopathologies would similarly benefit from the creation and validation of PRS and related tools. Collaborative efforts would also further allow for the inclusion of biosystems beyond genetics and neuroimaging, such as proteomics, metabolomics 423 or blood transcriptomics 424 , as well as potentially integrating some or all of these markers together in a multimodal framework to improve personalization and prediction.
Second, as alluded to above, alternate forms of classification and diagnosis may be useful for conceptualizing and testing potential biomarkers. This includes the investigation of intermediate phenotypes, which are related to the disorders but are more narrowly defined and putatively more proximal to the underlying neurobiological mechanisms (e.g., for anxiety disorders, the Research Domain Criteria “negative valence systems” traits 425 ).
Third, long‐term and cohort study designs are needed in order to evaluate the longitudinal course of disorder and remission/relapse after treatment, which may be more promising for biomarker development than diagnosis, being a clearer and more actionable target. Mechanistic confirmation may be achieved via experimental approaches that can demonstrate causality (by modifying the neural substrates thought to drive the disease states), such as neuromodulation techniques 426 or translational approaches incorporating animal models 427 .
Fourth, studies could include online remote acquisition of selected measures in a naturalistic setting using mobile health (mHealth) tools (e.g., ecological momentary assessment of physiological data 428 , 429 ). The identification of multimodal and multivariate signatures can be accomplished by means of mathematical modeling using artificial intelligence (e.g., machine learning, pattern recognition methods) in a systems biology approach 430 , 431 . Finally, a priori stratification approaches may be employed to clinically test preventive and therapeutic strategies individually tailored to the individual person's biological risk factor constellation.
In conclusion, although the general consensus is that we do not yet have clinically actionable biomarkers in psychiatry, considerable efforts and investments have fostered useful developments over the last couple of decades. We have reviewed some substantial advances made during this time, while also describing the additional work and complications that need to be addressed in order to accelerate and finalize the development and eventual roll out of candidate biomarkers into clinical settings. In doing so, some shifts in priorities may be necessary, particularly moving from diagnostic biomarker studies, which are currently overrepresented in the literature 84 , to targeting questions for which biomarkers may be most clinically actionable.
Recent examples highlight difficulties yielding highly accurate classifiers even when rich datasets and strong incentives for biomarker development are available 432 , and have specifically emphasized issues with external validation and robustness to sample‐specific factors. For a biomarker to be actionable, however, it will need to be clinically predictive at the individual‐person level. It will also need to be economically viable, which includes non‐prohibitive cost and the ability to deliver unique information that cannot be gleaned with traditional, less expensive alternatives.
These needs will evolve in parallel to the development of novel therapeutics and technologies, and so the menu of biomarker possibilities in psychiatry is likely to expand in the future. Even with the current state of affairs, though, suboptimal biomarker‐based predictions may still be helpful for high‐stakes applications if they improve upon what is possible when relying on clinical information alone, in ways that substantively decrease the individual and societal burden of mental illness. Setting specific benchmarks for well‐defined target applications that can be used today where possible, in conjunction with developing appropriate funding and partnership mechanisms for end‐to‐end biomarker development into the future, will be critical to ultimately reap the societal benefits of this critical scientific and clinical endeavor.
ACKNOWLEDGEMENTS
A. Abi‐Dargham is supported by grants from the US National Institute of Mental Health (NIMH) (R21MH125454) and the US National Institute on Drug Abuse (NIDA) (R61DA056423); S.J. Moeller by grants from NIDA (R01DA051420, R01DA049733, R21DA048196, R21DA051179 and R61DA056423); C. DeLorenzo by grants from the NIMH (R01MH114972, R01MH123093) and the US National Institute on Aging (RF1AG064245); K. Domschke by grants from the German Research Foundation (CRC‐TRR58, projects C02 and Z02; DO 1241/8‐1) and the German Federal Ministry of Education and Research (FKZ 01EE1402A, PROTECT‐AD, project P5); G. Horga by grants from the NIMH (R01MH117323 and R01MH114965); R. Kotov by a grant from the US National Institute of Occupational Safety and Health (U01OH011864); M.P. Paulus by the William K. Warren Foundation and the US National Institute of General Medical Sciences (1P20GM121312); G. Sanacora by the G.D. Gross and E.S. Gross Endowment and the Yale New Haven Health System, and J. Veenstra‐VanderWeele by the NIMH (R01MH114296) and the Simons Foundation. A. Abi‐Dargham and S.J. Moeller are joint first authors of the paper.
REFERENCES
- 1. Chang SM, Matchar DB, Smetana GW et al (eds). Methods guide for medical test reviews. Rockville: US Agency for Healthcare Research and Quality, 2012. [PubMed] [Google Scholar]
- 2. FDA‐NIH Biomarker Working Group . BEST (Biomarkers, EndpointS, and other Tools) resource. Silver Spring: US Food and Drug Administration, 2016. [PubMed] [Google Scholar]
- 3. Kane JM, Correll CU. The role of clozapine in treatment‐resistant schizophrenia. JAMA Psychiatry 2016;73:187‐8. [DOI] [PubMed] [Google Scholar]
- 4. Kong XZ, Zhen Z, Li X et al. Individual differences in impulsivity predict head motion during magnetic resonance imaging. PLoS One 2014;9:e104989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Miller GA, Chapman JP. Misunderstanding analysis of covariance. J Abnorm Psychol 2001;110:40‐8. [DOI] [PubMed] [Google Scholar]
- 6. Hastie T, Tibshirani R, Friedman R. The elements of statistical learning data mining, inference, and prediction, 2nd ed. New York: Springer, 2009. [Google Scholar]
- 7. Kim J‐H. Estimating classification error rate: repeated cross‐validation, repeated hold‐out and bootstrap. Comput Stat Data Anal 2009;53:3735‐45. [Google Scholar]
- 8. Alba AC, Agoritsas T, Walsh M et al. Discrimination and calibration of clinical prediction models: users' guides to the medical literature. JAMA 2017;318:1377‐84. [DOI] [PubMed] [Google Scholar]
- 9. Heagerty PJ, Zheng Y. Survival model predictive accuracy and ROC curves. Biometrics 2005;61:92‐105. [DOI] [PubMed] [Google Scholar]
- 10. First MB, Drevets WC, Carter C et al. Clinical applications of neuroimaging in psychiatric disorders. Am J Psychiatry 2018;175:915‐6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Abi‐Dargham A, Horga G. The search for imaging biomarkers in psychiatric disorders. Nat Med 2016;22:1248‐55. [DOI] [PubMed] [Google Scholar]
- 12. Belsher BE, Smolenski DJ, Pruitt LD et al. Prediction models for suicide attempts and deaths: a systematic review and simulation. JAMA Psychiatry 2019;76:642‐51. [DOI] [PubMed] [Google Scholar]
- 13. Liu X, Faes L, Kale AU et al. A comparison of deep learning performance against health‐care professionals in detecting diseases from medical imaging: a systematic review and meta‐analysis. Lancet Digit Health 2019;1:e271‐97. [DOI] [PubMed] [Google Scholar]
- 14. Koutsouleris N, Kambeitz‐Ilankovic L, Ruhrmann S et al. Prediction models of functional outcomes for individuals in the clinical high‐risk state for psychosis or with recent‐onset depression: a multimodal, multisite machine learning analysis. JAMA Psychiatry 2018;75:1156‐72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Kanner L. Autistic disturbances of affective contact. Nerv Child 1943;2:217‐50. [PubMed] [Google Scholar]
- 16. Weitlauf AS, McPheeters ML, Peters B et al. Therapies for children with autism spectrum disorder: behavioral interventions update. Rockville: US Agency for Healthcare Research and Quality, 2014. [PubMed] [Google Scholar]
- 17. Ameis SH, Kassee C, Corbett‐Dick P et al. Systematic review and guide to management of core and psychiatric symptoms in youth with autism. Acta Psychiatr Scand 2018;138:379‐400. [DOI] [PubMed] [Google Scholar]
- 18. Botha M, Hanlon J, Williams GL. Does language matter? Identity‐first versus person‐first language use in autism research: a response to Vivanti. J Autism Dev Disord 2023;53:870‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Volkow ND, Gordon JA, Koob GF. Choosing appropriate language to reduce the stigma around mental illness and substance use disorders. Neuropsychopharmacology 2021;46:2230‐2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Satterstrom FK, Kosmicki JA, Wang J et al. Large‐scale exome sequencing study implicates both developmental and functional changes in the neurobiology of autism. Cell 2020;180:568‐84.e523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Sanders SJ, Murtha MT, Gupta AR et al. De novo mutations revealed by whole‐exome sequencing are strongly associated with autism. Nature 2012;485:237‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Sanders SJ, He X, Willsey AJ et al. Insights into autism spectrum disorder genomic architecture and biology from 71 risk loci. Neuron 2015;87:1215‐33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Dong S, Walker MF, Carriero NJ et al. De novo insertions and deletions of predominantly paternal origin are associated with autism spectrum disorder. Cell Rep 2014;9:16‐23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Sanders SJ, Ercan‐Sencicek AG, Hus V et al. Multiple recurrent de novo CNVs, including duplications of the 7q11.23 Williams syndrome region, are strongly associated with autism. Neuron 2011;70:863‐85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Zhou X, Feliciano P, Wang T et al. Integrating de novo and inherited variants in over 42,607 autism cases identifies mutations in new moderate risk genes. Nat Genet 2022;54:1305‐19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Sebat J, Lakshmi B, Malhotra D et al. Strong association of de novo copy number mutations with autism. Science 2007;316:445‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Levy D, Ronemus M, Yamrom B et al. Rare de novo and transmitted copy‐number variation in autistic spectrum disorders. Neuron 2011;70:886‐97. [DOI] [PubMed] [Google Scholar]
- 28. Richards C, Jones C, Groves L et al. Prevalence of autism spectrum disorder phenomenology in genetic disorders: a systematic review and meta‐analysis. Lancet Psychiatry 2015;2:909‐16. [DOI] [PubMed] [Google Scholar]
- 29. Iossifov I, O'Roak BJ, Sanders SJ et al. The contribution of de novo coding mutations to autism spectrum disorder. Nature 2014;515:216‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Stessman HA, Xiong B, Coe BP et al. Nat Genet 2017;49:515‐26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Muhle RA, Reed HE, Vo LC et al. Clinical diagnostic genetic testing for individuals with developmental disorders. J Am Acad Child Adolesc Psychiatry 2017;56:910‐3. [DOI] [PubMed] [Google Scholar]
- 32. Arteche‐Lopez A, Gomez Rodriguez MJ, Sanchez Calvin MT et al. Towards a change in the diagnostic algorithm of autism spectrum disorders: evidence supporting whole exome sequencing as a first‐tier test. Genes 2021;12:560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Moreno‐De‐Luca D, Kavanaugh BC, Best CR et al. Clinical genetic testing in autism spectrum disorder in a large community‐based population sample. JAMA Psychiatry 2020;77:979‐81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Birnbaum R, Mahjani B, Loos RJF et al. Clinical characterization of copy number variants associated with neurodevelopmental disorders in a large‐scale multiancestry biobank. JAMA Psychiatry 2022;79:250‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Karalis V, Bateup HS. Current approaches and future directions for the treatment of mTORopathies. Dev Neurosci 2021;43:143‐58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Grove J, Ripke S, Als TD et al. Identification of common genetic risk variants for autism spectrum disorder. Nat Genet 2019;51:431‐4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Perkins DO, Olde Loohuis L, Barbee J et al. Polygenic risk score contribution to psychosis prediction in a target population of persons at clinical high risk. Am J Psychiatry 2020;177:155‐63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Zhang JP, Robinson D, Yu J et al. Schizophrenia polygenic risk score as a predictor of antipsychotic efficacy in first‐episode psychosis. Am J Psychiatry 2019;176:21‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Schain RJ, Freedman DX. Studies on 5‐hydroxyindole metabolism in autistic and other mentally retarded children. J Pediatrics 1961;58:315‐20. [DOI] [PubMed] [Google Scholar]
- 40. Bettelheim B. The empty fortress: infantile autism and the birth of the self. New York: Free Press, 1967. [Google Scholar]
- 41. Maenner MJ, Shaw KA, Bakian AV et al. Prevalence and characteristics of autism spectrum disorder among children aged 8 years – Autism and Developmental Disabilities Monitoring Network, 11 sites, United States, 2018. MMWR Surveill Summ 2021;70:1‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Gabriele S, Sacco R, Persico AM. Blood serotonin levels in autism spectrum disorder: a systematic review and meta‐analysis. Eur Neuropsychopharmacol 2014;24:919‐29. [DOI] [PubMed] [Google Scholar]
- 43. Mulder EJ, Anderson GM, Kema IP et al. Platelet serotonin levels in pervasive developmental disorders and mental retardation: diagnostic group differences, within‐group distribution, and behavioral correlates. J Am Acad Child Adolesc Psychiatry 2004;43:491‐9. [DOI] [PubMed] [Google Scholar]
- 44. Abney M, McPeek MS, Ober C. Broad and narrow heritabilities of quantitative traits in a founder population. Am J Hum Genet 2001;68:1302‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Shuffrey LC, Guter SJ, Delaney S et al. Is there sexual dimorphism of hyperserotonemia in autism spectrum disorder? Autism Res 2017;10:1417‐23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Piven J, Tsai G, Nehme E et al. Platelet serotonin, a possible marker for familial autism. J Autism Dev Disord 1991;21:51‐9. [DOI] [PubMed] [Google Scholar]
- 47. Muller CL, Anacker AMJ, Veenstra‐VanderWeele J. The serotonin system in autism spectrum disorder: from biomarker to animal models. Neuroscience 2016;321:24‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Zhao H, Zhang H, Liu S et al. Association of peripheral blood levels of cytokines with autism spectrum disorder: a meta‐analysis. Front Psychiatry 2021;12:670200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Chen L, Shi XJ, Liu H et al. Oxidative stress marker aberrations in children with autism spectrum disorder: a systematic review and meta‐analysis of 87 studies (N=9109). Transl Psychiatry 2021;11:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Andreo‐Martinez P, Rubio‐Aparicio M, Sanchez‐Meca J et al. A meta‐analysis of gut microbiota in children with autism. J Autism Dev Disord 2022;52:1374‐87. [DOI] [PubMed] [Google Scholar]
- 51. Bobrowski‐Khoury N, Ramaekers VT, Sequeira JM et al. Folate receptor alpha autoantibodies in autism spectrum disorders: diagnosis, treatment and prevention. J Pers Med 2021;11:710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Frye RE, Slattery J, Delhey L et al. Folinic acid improves verbal communication in children with autism and language impairment: a randomized double‐blind placebo‐controlled trial. Mol Psychiatry 2018;23:247‐56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Kong SW, Collins CD, Shimizu‐Motohashi Y et al. Characteristics and predictive value of blood transcriptome signature in males with autism spectrum disorders. PLoS One 2012;7:e49475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Pramparo T, Pierce K, Lombardo MV et al. Prediction of autism by translation and immune/inflammation coexpressed genes in toddlers from pediatric community practices. JAMA Psychiatry 2015;72:386‐94. [DOI] [PubMed] [Google Scholar]
- 55. Smith AM, Natowicz MR, Braas D et al. A metabolomics approach to screening for autism risk in the Children's Autism Metabolome Project. Autism Res 2020;13:1270‐85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Lainhart JE, Piven J, Wzorek M et al. Macrocephaly in children and adults with autism. J Am Acad Child Adolesc Psychiatry 1997;36:282‐90. [DOI] [PubMed] [Google Scholar]
- 57. Raznahan A, Wallace GL, Antezana L et al. Compared to what? Early brain overgrowth in autism and the perils of population norms. Biol Psychiatry 2013;74:563‐75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Lee JK, Andrews DS, Ozonoff S et al. Longitudinal evaluation of cerebral growth across childhood in boys and girls with autism spectrum disorder. Biol Psychiatry 2021;90:286‐94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Amaral DG, Li D, Libero L et al. In pursuit of neurophenotypes: the consequences of having autism and a big brain. Autism Res 2017;10:711‐22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Pua EPK, Bowden SC, Seal ML. Autism spectrum disorders: neuroimaging findings from systematic reviews. Res Autism Spectr Disord 2017;10:711‐22. [Google Scholar]
- 61. Rafiee F, Rezvani Habibabadi R, Motaghi M et al. Brain MRI in autism spectrum disorder: narrative review and recent advances. J Magn Reson Imaging 2022;55:1613‐24. [DOI] [PubMed] [Google Scholar]
- 62. Hull JV, Dokovna LB, Jacokes ZJ et al. Resting‐state functional connectivity in autism spectrum disorders: a review. Front Psychiatry 2016;7:205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Xie Y, Xu Z, Xia M et al. Alterations in connectome dynamics in autism spectrum disorder: a harmonized mega‐ and meta‐analysis study using the autism brain imaging data exchange dataset. Biol Psychiatry 2022;91:945‐55. [DOI] [PubMed] [Google Scholar]
- 64. Wolff JJ, Gu H, Gerig G et al. Differences in white matter fiber tract development present from 6 to 24 months in infants with autism. Am J Psychiatry 2012;169:589‐600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Hazlett HC, Gu H, Munsell BC et al. Early brain development in infants at high risk for autism spectrum disorder. Nature 2017;542:348‐51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Shen MD, Kim SH, McKinstry RC et al. Increased extra‐axial cerebrospinal fluid in high‐risk infants who later develop autism. Biol Psychiatry 2017;82:186‐93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Shen MD, Nordahl CW, Young GS et al. Early brain enlargement and elevated extra‐axial fluid in infants who develop autism spectrum disorder. Brain 2013;136:2825‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Shen MD, Nordahl CW, Li DD et al. Extra‐axial cerebrospinal fluid in high‐risk and normal‐risk children with autism aged 2‐4 years: a case‐control study. Lancet Psychiatry 2018;5:895‐904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. O'Reilly C, Lewis JD, Elsabbagh M. Is functional brain connectivity atypical in autism? A systematic review of EEG and MEG studies. PLoS One 2017;12:e0175870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Mash LE, Reiter MA, Linke AC et al. Multimodal approaches to functional connectivity in autism spectrum disorders: an integrative perspective. Dev Neurobiol 2018;78:456‐73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. McPartland J, Dawson G, Webb SJ et al. Event‐related brain potentials reveal anomalies in temporal processing of faces in autism spectrum disorder. J Child Psychol Psychiatry 2004;45:1235‐45. [DOI] [PubMed] [Google Scholar]
- 72. Kang E, Keifer CM, Levy EJ et al. Atypicality of the N170 event‐related potential in autism spectrum disorder: a meta‐analysis. Biol Psychiatry Cogn Neurosci Neuroimaging 2018;3:657‐66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. McPartland JC, Bernier RA, Jeste SS et al. The Autism Biomarkers Consortium for Clinical Trials (ABC‐CT): scientific context, study design, and progress toward biomarker qualification. Front Integr Neurosci 2020;14:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Kala S, Rolison MJ, Trevisan DA et al. Brief report: Preliminary evidence of the N170 as a biomarker of response to treatment in autism spectrum disorder. Front Psychiatry 2021;12:709382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Ajram LA, Pereira AC, Durieux AMS et al. The contribution of [1H] magnetic resonance spectroscopy to the study of excitation‐inhibition in autism. Prog Neuropsychopharmacol Biol Psychiatry 2019;89:236‐44. [DOI] [PubMed] [Google Scholar]
- 76. Ford TC, Crewther DP. A comprehensive review of the (1)H‐MRS metabolite spectrum in autism spectrum disorder. Front Mol Neurosci 2016;9:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Tan Z, Wei H, Song X et al. Positron emission tomography in the neuroimaging of autism spectrum disorder: a review. Front Neurosci 2022;16:806876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Oztan O, Garner JP, Constantino JN et al. Neonatal CSF vasopressin concentration predicts later medical record diagnoses of autism spectrum disorder. Proc Natl Acad Sci USA 2020;117:10609‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Oztan O, Garner JP, Partap S et al. Cerebrospinal fluid vasopressin and symptom severity in children with autism. Ann Neurol 2018;84:611‐5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Parker KJ, Oztan O, Libove RA et al. A randomized placebo‐controlled pilot trial shows that intranasal vasopressin improves social deficits in children with autism. Sci Transl Med 2019;11:eaau7356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Charlson FJ, Ferrari AJ, Santomauro DF et al. Global epidemiology and burden of schizophrenia: findings from the Global Burden of Disease Study 2016. Schizophr Bull 2018;44:1195‐203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Rubio JM, Taipale H, Tanskanen A et al. Long‐term continuity of antipsychotic treatment for schizophrenia: a nationwide study. Schizophr Bull 2021;47:1611‐20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Meehan AJ, Lewis SJ, Fazel S et al. Clinical prediction models in psychiatry: a systematic review of two decades of progress and challenges. Mol Psychiatry 2022;27:2700‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Kraguljac NV, McDonald WM, Widge AS et al. Neuroimaging biomarkers in schizophrenia. Am J Psychiatry 2021;178:509‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Kahn RS, Sommer IE, Murray RM et al. Schizophrenia. Nat Rev Dis Primers 2015;1:15067. [DOI] [PubMed] [Google Scholar]
- 86. Sehatpour P, Javitt DC, De Baun HM et al. Mismatch negativity as an index of target engagement for excitation/inhibition‐based treatment development: a double‐blind, placebo‐controlled, randomized, single‐dose cross‐over study of the serotonin type‐3 receptor antagonist CVN058. Neuropsychopharmacology 2022;47:711‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Thompson E, Millman ZB, Okuzawa N et al. Evidence‐based early interventions for individuals at clinical high risk for psychosis: a review of treatment components. J Nerv Ment Dis 2015;203:342‐51. [DOI] [PubMed] [Google Scholar]
- 88. Addington J, Addington D, Abidi S et al. Canadian treatment guidelines for individuals at clinical high risk of psychosis. Can J Psychiatry 2017;62:656‐61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. US National Institute of Mental Health . Accelerating Medicines Partnership® Program – Schizophrenia (AMP® SCZ). Bethesda: US National Institute of Mental Health, 2022. [Google Scholar]
- 90. Javitt DC. D‐serine for the schizophrenia prodrome. clinicaltrials.gov/ct2/show/NCT00826202.
- 91. Ingelheim Boehringer. A phase II randomised, double‐blind, placebo‐controlled study to evaluate the efficacy, safety, and tolerability of orally administered BI 409306 during a 52‐week treatment period as an early intervention in patients with attenuated psychosis. clinicaltrials.gov/ct2/show/NCT03230097.
- 92. Cannon TD, Yu C, Addington J et al. An individualized risk calculator for research in prodromal psychosis. Am J Psychiatry 2016;173:980‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Zhang T, Li H, Tang Y et al. Validating the predictive accuracy of the NAPLS‐2 psychosis risk calculator in a clinical high‐risk sample from the SHARP (Shanghai At Risk for Psychosis) Program. Am J Psychiatry 2018;175:906‐8. [DOI] [PubMed] [Google Scholar]
- 94. Carrión RE, Cornblatt BA, Burton CZ et al. Personalized prediction of psychosis: external validation of the NAPLS‐2 psychosis risk calculator with the EDIPPP Project. Am J Psychiatry 2016;173:989‐96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Miller TJ, McGlashan TH, Rosen JL et al. Prodromal assessment with the Structured Interview for Prodromal Syndromes and the Scale of Prodromal Symptoms: predictive validity, interrater reliability, and training to reliability. Schizophr Bull 2003;29:703‐15. [DOI] [PubMed] [Google Scholar]
- 96. Koutsouleris N, Dwyer DB, Degenhardt F et al. Multimodal machine learning workflows for prediction of psychosis in patients with clinical high‐risk syndromes and recent‐onset depression. JAMA Psychiatry 2021;78:195‐209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Kegeles LS, Ciarleglio A, León‐Ortiz P et al. An imaging‐based risk calculator for prediction of conversion to psychosis in clinical high‐risk individuals using glutamate 1H MRS. Schizophr Res 2020;226:70‐3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Chung Y, Addington J, Bearden CE et al. Use of machine learning to determine deviance in neuroanatomical maturity associated with future psychosis in youths at clinically high risk. JAMA Psychiatry 2018;75:960‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Bodatsch M, Ruhrmann S, Wagner M et al. Prediction of psychosis by mismatch negativity. Biol Psychiatry 2011;69:959‐66. [DOI] [PubMed] [Google Scholar]
- 100. Perez VB, Woods SW, Roach BJ et al. Automatic auditory processing deficits in schizophrenia and clinical high‐risk patients: forecasting psychosis risk with mismatch negativity. Biol Psychiatry 2014;75:459‐69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Fryer SL, Roach BJ, Hamilton HK et al. Deficits in auditory predictive coding in individuals with the psychosis risk syndrome: prediction of conversion to psychosis. J Abnorm Psychol 2020;129:599‐611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Cao H, Chén OY, Chung Y et al. Cerebello‐thalamo‐cortical hyperconnectivity as a state‐independent functional neural signature for psychosis prediction and characterization. Nat Commun 2018;9:3836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Mizrahi R, Addington J, Rusjan PM et al. Increased stress‐induced dopamine release in psychosis. Biol Psychiatry 2012;71:561‐7. [DOI] [PubMed] [Google Scholar]
- 104. Howes OD, Montgomery AJ, Asselin MC et al. Elevated striatal dopamine function linked to prodromal signs of schizophrenia. Arch Gen Psychiatry 2009;66:13‐20. [DOI] [PubMed] [Google Scholar]
- 105. Howes OD, Bose SK, Turkheimer F et al. Dopamine synthesis capacity before onset of psychosis: a prospective [18F]‐DOPA PET imaging study. Am J Psychiatry 2011;168:1311‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Meyer‐Lindenberg A, Miletich RS, Kohn PD et al. Reduced prefrontal activity predicts exaggerated striatal dopaminergic function in schizophrenia. Nat Neurosci 2002;5:267‐71. [DOI] [PubMed] [Google Scholar]
- 107. Fusar‐Poli P, Howes OD, Allen P et al. Abnormal frontostriatal interactions in people with prodromal signs of psychosis: a multimodal imaging study. Arch Gen Psychiatry 2010;67:683‐91. [DOI] [PubMed] [Google Scholar]
- 108. Fusar‐Poli P, Howes OD, Allen P et al. Abnormal prefrontal activation directly related to pre‐synaptic striatal dopamine dysfunction in people at clinical high risk for psychosis. Mol Psychiatry 2011;16:67‐75. [DOI] [PubMed] [Google Scholar]
- 109. Schifani C, Tseng HH, Kenk M et al. Cortical stress regulation is disrupted in schizophrenia but not in clinical high risk for psychosis. Brain 2018;141:2213‐24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Howes O, Bose S, Turkheimer F et al. Progressive increase in striatal dopamine synthesis capacity as patients develop psychosis: a PET study. Mol Psychiatry 2011;16:885‐6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Egerton A, Chaddock CA, Winton‐Brown TT et al. Presynaptic striatal dopamine dysfunction in people at ultra‐high risk for psychosis: findings in a second cohort. Biol Psychiatry 2013;74:106‐12. [DOI] [PubMed] [Google Scholar]
- 112. Horga G, Wengler K, Cassidy CM. Neuromelanin‐sensitive magnetic resonance imaging as a proxy marker for catecholamine function in psychiatry. JAMA Psychiatry 2021;78:788‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Cassidy CM, Zucca FA, Girgis RR et al. Neuromelanin‐sensitive MRI as a noninvasive proxy measure of dopamine function in the human brain. Proc Natl Acad Sci USA 2019;116:5108‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Whiting D, Gulati G, Geddes JR et al. Association of schizophrenia spectrum disorders and violence perpetration in adults and adolescents from 15 countries: a systematic review and meta‐analysis. JAMA Psychiatry 2022;79:120‐32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Olfson M, Stroup TS, Huang C et al. Suicide risk in Medicare patients with schizophrenia across the life span. JAMA Psychiatry 2021;78:876‐85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Singh JP, Serper M, Reinharth J et al. Structured assessment of violence risk in schizophrenia and other psychiatric disorders: a systematic review of the validity, reliability, and item content of 10 available instruments. Schizophr Bull 2011;37:899‐912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. SanSegundo MS, Ferrer‐Cascales R, Bellido JH et al. Prediction of violence, suicide behaviors and suicide ideation in a sample of institutionalized offenders with schizophrenia and other psychosis. Front Psychol 2018;9:1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Widmayer S, Borgwardt S, Lang UE et al. Functional neuroimaging correlates of aggression in psychosis: a systematic review with recommendations for future research. Front Psychiatry 2019;9:777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Arango C, Calcedo Barba A, Gonzalez S et al. Violence in inpatients with schizophrenia: a prospective study. Schizophr Bull 1999;25:493‐503. [DOI] [PubMed] [Google Scholar]
- 120. Correll CU, Howes OD. Treatment‐resistant schizophrenia: definition, predictors, and therapy options. J Clin Psychiatry 2021;82:MY20096AH1C. [DOI] [PubMed] [Google Scholar]
- 121. Novartis Pharmaceuticals . Clozaril (clozapine). https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/019758s062lbl.pdf.
- 122. Brannan SK, Sawchak S, Miller AC et al. Muscarinic cholinergic receptor agonist and peripheral antagonist for schizophrenia. N Engl J Med 2021;384:717‐26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Li A, Zalesky A, Yue W et al. A neuroimaging biomarker for striatal dysfunction in schizophrenia. Nat Med 2020;26:558‐65. [DOI] [PubMed] [Google Scholar]
- 124. Sarpal DK, Argyelan M, Robinson DG et al. Baseline striatal functional connectivity as a predictor of response to antipsychotic drug treatment. Am J Psychiatry 2016;173:69‐77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Zou QH, Zhu CZ, Yang Y et al. An improved approach to detection of amplitude of low‐frequency fluctuation (ALFF) for resting‐state fMRI: fractional ALFF. J Neurosci Methods 2008;172:137‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Di Martino A, Scheres A, Margulies DS et al. Functional connectivity of human striatum: a resting state FMRI study. Cereb Cortex 2008;18:2735‐47. [DOI] [PubMed] [Google Scholar]
- 127. Sarpal DK, Robinson DG, Fales C et al. Relationship between duration of untreated psychosis and intrinsic corticostriatal connectivity in patients with early phase schizophrenia. Neuropsychopharmacology 2017;42:2214‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Rubio JM, Lencz T, Barber A et al. Striatal functional connectivity in psychosis relapse: a hypothesis generating study. Schizophr Res 2022;243:342‐8. [DOI] [PubMed] [Google Scholar]
- 129. Blair Thies M, DeRosse P, Sarpal DK et al. Interaction of cannabis use disorder and striatal connectivity in antipsychotic treatment response. Schizophr Bull Open 2020;1:sgaa014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Cao B, Cho RY, Chen D et al. Treatment response prediction and individualized identification of first‐episode drug‐naïve schizophrenia using brain functional connectivity. Mol Psychiatry 2020;25:906‐13. [DOI] [PubMed] [Google Scholar]
- 131. Kraguljac NV, White DM, Hadley N et al. Aberrant hippocampal connectivity in unmedicated patients with schizophrenia and effects of antipsychotic medication: a longitudinal resting state functional MRI study. Schizophr Bull 2016;42:1046‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Blessing EM, Murty VP, Zeng B et al. Anterior hippocampal‐cortical functional connectivity distinguishes antipsychotic naïve first‐episode psychosis patients from controls and may predict response to second‐generation antipsychotic treatment. Schizophr Bull 2020;46:680‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Egerton A, Brugger S, Raffin M et al. Anterior cingulate glutamate levels related to clinical status following treatment in first‐episode schizophrenia. Neuropsychopharmacology 2012;37:2515‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Mouchlianitis E, Bloomfield MA, Law V et al. Treatment‐resistant schizophrenia patients show elevated anterior cingulate cortex glutamate compared to treatment‐responsive. Schizophr Bull 2016;42:744‐52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Iwata Y, Nakajima S, Plitman E et al. Glutamatergic neurometabolite levels in patients with ultra‐treatment‐resistant schizophrenia: a cross‐sectional 3T proton magnetic resonance spectroscopy study. Biol Psychiatry 2019;85:596‐605. [DOI] [PubMed] [Google Scholar]
- 136. Egerton A, Murphy A, Donocik J et al. Dopamine and glutamate in antipsychotic‐responsive compared with antipsychotic‐nonresponsive psychosis: a multicenter positron emission tomography and magnetic resonance spectroscopy study (STRATA). Schizophr Bull 2021;47:505‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. van de Giessen E, van der Pluijm M, Booij J et al. Neuromelanin MRI as biomarker for treatment resistance in first episode schizophrenia. Biol Psychiatry 2022;91(Suppl. 9):S61. [Google Scholar]
- 138. Veronese M, Santangelo B, Jauhar S et al. A potential biomarker for treatment stratification in psychosis: evaluation of an [18F] FDOPA PET imaging approach. Neuropsychopharmacology 2021;46:1122‐32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Abi‐Dargham A, Rodenhiser J, Printz D et al. Increased baseline occupancy of D2 receptors by dopamine in schizophrenia. Proc Natl Acad Sci USA 2000;97:8104‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Jauhar S, Veronese M, Nour MM et al. Determinants of treatment response in first‐episode psychosis: an 18F‐DOPA PET study. Mol Psychiatry 2019;24:1502‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Robinson D, Woerner MG, Alvir JM et al. Predictors of relapse following response from a first episode of schizophrenia or schizoaffective disorder. Arch Gen Psychiatry 1999;56:241‐7. [DOI] [PubMed] [Google Scholar]
- 142. Taipale H, Tanskanen A, Correll CU et al. Real‐world effectiveness of antipsychotic doses for relapse prevention in patients with first‐episode schizophrenia in Finland: a nationwide, register‐based cohort study. Lancet Psychiatry 2022;9:271‐9. [DOI] [PubMed] [Google Scholar]
- 143. Takeuchi H, Siu C, Remington G et al. Does relapse contribute to treatment resistance? Antipsychotic response in first‐ vs. second‐episode schizophrenia. Neuropsychopharmacology 2019;44:1036‐42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Leucht S, Tardy M, Komossa K et al. Antipsychotic drugs versus placebo for relapse prevention in schizophrenia: a systematic review and meta‐analysis. Lancet 2012;379:2063‐71. [DOI] [PubMed] [Google Scholar]
- 145. Alvarez‐Jimenez M, Priede A, Hetrick SE et al. Risk factors for relapse following treatment for first episode psychosis: a systematic review and meta‐analysis of longitudinal studies. Schizophr Res 2012;139:116‐28. [DOI] [PubMed] [Google Scholar]
- 146. Rubio JM, Schoretsanitis G, John M et al. Psychosis relapse during treatment with long‐acting injectable antipsychotics in individuals with schizophrenia‐spectrum disorders: an individual participant data meta‐analysis. Lancet Psychiatry 2020;7:749‐61. [DOI] [PubMed] [Google Scholar]
- 147. Rubio JM, Malhotra AK, Kane JM. Towards a framework to develop neuroimaging biomarkers of relapse in schizophrenia. Behav Brain Res 2021;402:113099. [DOI] [PubMed] [Google Scholar]
- 148. Herz MI, Lamberti JS, Mintz J et al. A program for relapse prevention in schizophrenia: a controlled study. Arch Gen Psychiatry 2000;57:277‐83. [DOI] [PubMed] [Google Scholar]
- 149. Light GA, Braff DL. Mismatch negativity deficits are associated with poor functioning in schizophrenia patients. Arch Gen Psychiatry 2005;62:127‐36. [DOI] [PubMed] [Google Scholar]
- 150. Light GA, Swerdlow NR, Rissling AJ et al. Characterization of neurophysiologic and neurocognitive biomarkers for use in genomic and clinical outcome studies of schizophrenia. PLoS One 2012;7:e39434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Friedman T, Sehatpour P, Dias E et al. Differential relationships of mismatch negativity and visual P1 deficits to premorbid characteristics and functional outcome in schizophrenia. Biol Psychiatry 2012;71:521‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Hochberger WC, Joshi YB, Thomas ML et al. Neurophysiologic measures of target engagement predict response to auditory‐based cognitive training in treatment refractory schizophrenia. Neuropsychopharmacology 2019;44:606‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Biagianti B, Roach BJ, Fisher M et al. Trait aspects of auditory mismatch negativity predict response to auditory training in individuals with early illness schizophrenia. Neuropsychiatr Electrophysiol 2017;3:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Medalia A, Saperstein AM, Qian M et al. Impact of baseline early auditory processing on response to cognitive remediation for schizophrenia. Schizophr Res 2019;208:397‐405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Kim HK, Blumberger DM, Daskalakis ZJ. Neurophysiological biomarkers in schizophrenia – P50, mismatch negativity, and TMS‐EMG and TMS‐EEG. Front Psychiatry 2020;11:795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Qi W, Marx J, Zingman M et al. Hippocampal subfield volumes predict disengagement from maintenance treatment in first episode schizophrenia. Schizophr Bull 2023;49:34‐42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Engl J, Rettenbacher M, Fleischhacker WW et al. Atypical antipsychotic drugs directly impair insulin action in adipocytes: effects on glucose transport, lipogenesis, and antilipolysis. Neuropsychopharmacology 2007;32:2431‐2. [DOI] [PubMed] [Google Scholar]
- 158. Guinart D, Taipale H, Rubio JM et al. Risk factors, incidence, and outcomes of neuroleptic malignant syndrome on long‐acting injectable vs oral antipsychotics in a nationwide schizophrenia cohort. Schizophr Bull 2021;47:1621‐30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Mathews J, Newcomer JW, Mathews JR et al. Neural correlates of weight gain with olanzapine. Arch Gen Psychiatry 2012;69:1226‐37. [DOI] [PubMed] [Google Scholar]
- 160. Kim SF, Huang AS, Snowman AM et al. Antipsychotic drug‐induced weight gain mediated by histamine H1 receptor‐linked activation of hypothalamic AMP‐kinase. Proc Natl Acad Sci USA 2007;104:3456‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Nielsen M, Rostrup E, Wulff S et al. Striatal reward activity and antipsychotic‐associated weight change in patients with schizophrenia undergoing initial treatment. JAMA Psychiatry 2016;73:121‐8. [DOI] [PubMed] [Google Scholar]
- 162. Homan P, Argyelan M, Fales CL et al. Striatal volume and functional connectivity correlate with weight gain in early‐phase psychosis. Neuropsychopharmacology 2019;44:1948‐54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Stegmayer K, Walther S, van Harten P. Tardive dyskinesia associated with atypical antipsychotics: prevalence, mechanisms and management strategies. CNS Drugs 2018;32:135‐47. [DOI] [PubMed] [Google Scholar]
- 164. Trubetskoy V, Pardiñas AF, Qi T et al. Mapping genomic loci implicates genes and synaptic biology in schizophrenia. Nature 2022;604:502‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Kessler RC, Petukhova M, Sampson NA et al. Twelve‐month and lifetime prevalence and lifetime morbid risk of anxiety and mood disorders in the United States. Int J Methods Psychiatr Res 2012;21:169‐84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Penninx BW, Pine DS, Holmes EA et al. Anxiety disorders. Lancet 2021;397:914‐27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Wittchen HU, Jacobi F, Rehm J et al. The size and burden of mental disorders and other disorders of the brain in Europe 2010. Eur Neuropsychopharmacol 2011;21:655‐79. [DOI] [PubMed] [Google Scholar]
- 168. Baxter AJ, Vos T, Scott KM et al. The global burden of anxiety disorders in 2010. Psychol Med 2014;44:2363‐74. [DOI] [PubMed] [Google Scholar]
- 169. Olesen J, Gustavsson A, Svensson M et al. The economic cost of brain disorders in Europe. Eur J Neurol 2012;19:155‐62. [DOI] [PubMed] [Google Scholar]
- 170. Meier SM, Petersen L, Mattheisen M et al. Secondary depression in severe anxiety disorders: a population‐based cohort study in Denmark. Lancet Psychiatry 2015;2:515‐23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Grant BF, Saha TD, Ruan WJ et al. Epidemiology of DSM‐5 drug use disorder: results from the National Epidemiologic Survey on Alcohol and Related Conditions‐III. JAMA Psychiatry 2016;73:39‐47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Bystritsky A. Treatment‐resistant anxiety disorders. Mol Psychiatry 2006;11:805‐14. [DOI] [PubMed] [Google Scholar]
- 173. Loerinc AG, Meuret AE, Twohig MP et al. Response rates for CBT for anxiety disorders: need for standardized criteria. Clin Psychol Rev 2015;42:72‐82. [DOI] [PubMed] [Google Scholar]
- 174. Taylor S, Abramowitz JS, McKay D. Non‐adherence and non‐response in the treatment of anxiety disorders. J Anxiety Disord 2012;26:583‐9. [DOI] [PubMed] [Google Scholar]
- 175. Solis EC, van Hemert AM, Carlier IVE et al. The 9‐year clinical course of depressive and anxiety disorders: new NESDA findings. J Affect Disord 2021;295:1269‐79. [DOI] [PubMed] [Google Scholar]
- 176. Bandelow B, Baldwin D, Abelli M et al. Biological markers for anxiety disorders, OCD and PTSD – a consensus statement. Part I: Neuroimaging and genetics. World J Biol Psychiatry 2016;17:321‐65. [DOI] [PubMed] [Google Scholar]
- 177. Bandelow B, Baldwin D, Abelli M et al. Biological markers for anxiety disorders, OCD and PTSD: a consensus statement. Part II: Neurochemistry, neurophysiology and neurocognition. World J Biol Psychiatry 2017;18:162‐214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Cosci F, Mansueto G. Biological and clinical markers to differentiate the type of anxiety disorders. Adv Exp Med Biol 2020;1191:197‐218. [DOI] [PubMed] [Google Scholar]
- 179. Deckert J, Erhardt A. Predicting treatment outcome for anxiety disorders with or without comorbid depression using clinical, imaging and (epi)genetic data. Curr Opin Psychiatry 2019;32:1‐6. [DOI] [PubMed] [Google Scholar]
- 180. Maron E, Lan CC, Nutt D. Imaging and genetic approaches to inform biomarkers for anxiety disorders, obsessive‐compulsive disorders, and PSTD. Curr Top Behav Neurosci 2018;40:219‐92. [DOI] [PubMed] [Google Scholar]
- 181. Vismara M, Girone N, Cirnigliaro G et al. Peripheral biomarkers in DSM‐5 anxiety disorders: an updated overview. Brain Sci 2020;10:564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Howe AS, Buttenschøn HN, Bani‐Fatemi A et al. Candidate genes in panic disorder: meta‐analyses of 23 common variants in major anxiogenic pathways. Mol Psychiatry 2016;21:665‐79. [DOI] [PubMed] [Google Scholar]
- 183. Gottschalk MG, Domschke K. Genetics of generalized anxiety disorder and related traits. Dialogues Clin Neurosci 2017;19:159‐68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Gottschalk MG, Domschke K. Novel developments in genetic and epigenetic mechanisms of anxiety. Curr Opin Psychiatry 2016;29:32‐8. [DOI] [PubMed] [Google Scholar]
- 185. Smoller JW. The genetics of stress‐related disorders: PTSD, depression, and anxiety disorders. Neuropsychopharmacology 2016;41:297‐319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Scherf‐Clavel M, Weber H, Deckert J et al. The role of pharmacogenetics in the treatment of anxiety disorders and the future potential for targeted therapeutics. Expert Opin Drug Metab Toxicol 2021;17:1249‐60. [DOI] [PubMed] [Google Scholar]
- 187. Levey DF, Gelernter J, Polimanti R et al. Reproducible genetic risk loci for anxiety: results from ∼200,000 participants in the Million Veteran Program. Am J Psychiatry 2020;177:223‐32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Asselmann E, Hertel J, Schmidt CO et al. Interplay between RGS2 and childhood adversities in predicting anxiety and depressive disorders: findings from a general population sample. Depress Anxiety 2018;35:1104‐13. [DOI] [PubMed] [Google Scholar]
- 189. Li C, Liang X, Cheng S et al. A multi‐environments‐gene interaction study of anxiety, depression and self‐harm in the UK Biobank cohort. J Psychiatr Res 2022;147:59‐66. [DOI] [PubMed] [Google Scholar]
- 190. Schiele MA, Herzog K, Kollert L et al. Extending the vulnerability‐stress model of mental disorders: three‐dimensional NPSR1 × environment × coping interaction study in anxiety. Br J Psychiatry 2020;217:645‐50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Domschke K. Prevention in psychiatry: a role for epigenetics? World Psychiatry 2021;20:227‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Schiele MA, Gottschalk MG, Domschke K. The applied implications of epigenetics in anxiety, affective and stress‐related disorders – A review and synthesis on psychosocial stress, psychotherapy and prevention. Clin Psychol Rev 2020;77:101830. [DOI] [PubMed] [Google Scholar]
- 193. Schiele MA, Domschke K. Epigenetics at the crossroads between genes, environment and resilience in anxiety disorders. Genes Brain Behav 2018;17:e12423. [DOI] [PubMed] [Google Scholar]
- 194. Iurato S, Carrillo‐Roa T, Arloth J et al. DNA methylation signatures in panic disorder. Transl Psychiatry 2017;7:1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Shimada‐Sugimoto M, Otowa T, Miyagawa T et al. Epigenome‐wide association study of DNA methylation in panic disorder. Clin Epigenetics 2017;9:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Ziegler C, Grundner‐Culemann F, Schiele MA et al. The DNA methylome in panic disorder: a case‐control and longitudinal psychotherapy‐epigenetic study. Transl Psychiatry 2019;9:314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Wiegand A, Kreifelts B, Munk MHJ et al. DNA methylation differences associated with social anxiety disorder and early life adversity. Transl Psychiatry 2021;11:104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Jonas KG, Lencz T, Li K et al. Schizophrenia polygenic risk score and 20‐year course of illness in psychotic disorders. Transl Psychiatry 2019;9:300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Bakulski KM, Halladay A, Hu VW et al. Epigenetic research in neuropsychiatric disorders: the “tissue issue”. Curr Behav Neurosci Rep 2016;3:264‐74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Lebow MA, Chen A. Overshadowed by the amygdala: the bed nucleus of the stria terminalis emerges as key to psychiatric disorders. Mol Psychiatry 2016;21:450‐63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Etkin A, Wager TD. Functional neuroimaging of anxiety: a meta‐analysis of emotional processing in PTSD, social anxiety disorder, and specific phobia. Am J Psychiatry 2007;164:1476‐88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Hyde J, Ryan KM, Waters AM. Psychophysiological markers of fear and anxiety. Curr Psychiatry Rep 2019;21:56. [DOI] [PubMed] [Google Scholar]
- 203. Meyer A, Nelson B, Perlman G et al. A neural biomarker, the error‐related negativity, predicts the first onset of generalized anxiety disorder in a large sample of adolescent females. J Child Psychol Psychiatry 2018;59:1162‐70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Domschke K, Stevens S, Pfleiderer B et al. Interoceptive sensitivity in anxiety and anxiety disorders: an overview and integration of neurobiological findings. Clin Psychol Rev 2010;30:1‐11. [DOI] [PubMed] [Google Scholar]
- 205. Robinaugh DJ, Ward MJ, Toner ER et al. Assessing vulnerability to panic: a systematic review of psychological and physiological responses to biological challenges as prospective predictors of panic attacks and panic disorder. Gen Psychiatr 2019;32:e100140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Vollmer LL, Strawn JR, Sah R. Acid‐base dysregulation and chemosensory mechanisms in panic disorder: a translational update. Transl Psychiatry 2015;5:e572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Kaplan JS, Arnkoff DB, Glass CR et al. Avoidant coping in panic disorder: a yohimbine biological challenge study. Anxiety Stress Coping 2012;25:425‐42. [DOI] [PubMed] [Google Scholar]
- 208. Van Veen JF, Van der Wee NJ, Fiselier J et al. Behavioural effects of rapid intravenous administration of meta‐chlorophenylpiperazine (m‐CPP) in patients with generalized social anxiety disorder, panic disorder and healthy controls. Eur Neuropsychopharmacol 2007;17:637‐42. [DOI] [PubMed] [Google Scholar]
- 209. Bradwejn J, Koszycki D, Shriqui C. Enhanced sensitivity to cholecystokinin tetrapeptide in panic disorder. Clinical and behavioral findings. Arch Gen Psychiatry 1991;48:603‐10. [DOI] [PubMed] [Google Scholar]
- 210. Bradwejn J, Koszycki D, Annable L et al. A dose‐ranging study of the behavioral and cardiovascular effects of CCK‐tetrapeptide in panic disorder. Biol Psychiatry 1992;32:903‐12. [DOI] [PubMed] [Google Scholar]
- 211. Schiele MA, Reif A, Lin J et al. Therapygenetic effects of 5‐HTTLPR on cognitive‐behavioral therapy in anxiety disorders: a meta‐analysis. Eur Neuropsychopharmacol 2021;44:105‐20. [DOI] [PubMed] [Google Scholar]
- 212. Lueken U, Zierhut KC, Hahn T et al. Neurobiological markers predicting treatment response in anxiety disorders: a systematic review and implications for clinical application. Neurosci Biobehav Rev 2016;66:143‐62. [DOI] [PubMed] [Google Scholar]
- 213. Rayner C, Coleman JRI, Purves KL et al. A genome‐wide association meta‐analysis of prognostic outcomes following cognitive behavioural therapy in individuals with anxiety and depressive disorders. Transl Psychiatry 2019;9:150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Tomasi J, Lisoway AJ, Zai CC et al. Towards precision medicine in generalized anxiety disorder: review of genetics and pharmaco(epi)genetics. J Psychiatr Res 2019;119:33‐47. [DOI] [PubMed] [Google Scholar]
- 215. Bradley P, Shiekh M, Mehra V et al. Improved efficacy with targeted pharmacogenetic‐guided treatment of patients with depression and anxiety: a randomized clinical trial demonstrating clinical utility. J Psychiatr Res 2018;96:100‐7. [DOI] [PubMed] [Google Scholar]
- 216. Jung J, Tawa EA, Muench C et al. Genome‐wide association study of treatment response to venlafaxine XR in generalized anxiety disorder. Psychiatry Res 2017;254:8‐11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Picó‐Pérez M, Fullana MA, Albajes‐Eizagirre A et al. Neural predictors of cognitive‐behavior therapy outcome in anxiety‐related disorders: a meta‐analysis of task‐based fMRI studies. Psychol Med 2022; doi: 10.1017/S0033291721005444. [DOI] [PubMed] [Google Scholar]
- 218. Whitfield‐Gabrieli S, Ghosh SS, Nieto‐Castanon A et al. Brain connectomics predict response to treatment in social anxiety disorder. Mol Psychiatry 2016;21:680‐5. [DOI] [PubMed] [Google Scholar]
- 219. Frick A, Engman J, Alaie I et al. Neuroimaging, genetic, clinical, and demographic predictors of treatment response in patients with social anxiety disorder. J Affect Disord 2020;261:230‐7. [DOI] [PubMed] [Google Scholar]
- 220. Van der Linden G, van Heerden B, Warwick J et al. Functional brain imaging and pharmacotherapy in social phobia: single photon emission computed tomography before and after treatment with the selective serotonin reuptake inhibitor citalopram. Prog Neuropsychopharmacol Biol Psychiatry 2000;24:419‐38. [DOI] [PubMed] [Google Scholar]
- 221. Klumpp H, Fitzgerald JM. Neuroimaging predictors and mechanisms of treatment response in social anxiety disorder: an overview of the amygdala. Curr Psychiatry Rep 2018;20:89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Maron E, Nutt D. Biological predictors of pharmacological therapy in anxiety disorders. Dialogues Clin Neurosci 2015;17:305‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Perna G, Alciati A, Sangiorgio E et al. Personalized clinical approaches to anxiety disorders. Adv Exp Med Biol 2020;1191:489‐521. [DOI] [PubMed] [Google Scholar]
- 224. Ball TM, Stein MB, Ramsawh HJ et al. Single‐subject anxiety treatment outcome prediction using functional neuroimaging. Neuropsychopharmacology 2014;39:1254‐61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Frick A, Engman J, Wahlstedt K et al. Anterior cingulate cortex activity as a candidate biomarker for treatment selection in social anxiety disorder. BJPsych Open 2018;4:157‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Nelson BD, Jackson F, Amir N et al. Attention bias modification reduces neural correlates of response monitoring. Biol Psychol 2017;129:103‐10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Blevins CA, Weathers FW, Davis MT et al. The Posttraumatic Stress Disorder Checklist for DSM‐5 (PCL‐5): development and initial psychometric evaluation. J Trauma Stress 2015;28:489‐98. [DOI] [PubMed] [Google Scholar]
- 228. Hoskins M, Pearce J, Bethell A et al. Pharmacotherapy for post‐traumatic stress disorder: systematic review and meta‐analysis. Br J Psychiatry 2015;206:93‐100. [DOI] [PubMed] [Google Scholar]
- 229. Polimanti R, Wendt FR. Posttraumatic stress disorder: from gene discovery to disease biology. Psychol Med 2021;51:2178‐88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230. Stein MB, Levey DF, Cheng Z et al. Genome‐wide association analyses of post‐traumatic stress disorder and its symptom subdomains in the Million Veteran Program. Nat Genet 2021;53:174‐84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Tamman AJF, Wendt FR, Pathak GA et al. Attachment style moderates polygenic risk for incident posttraumatic stress in U.S. military veterans: a 7‐year, nationally representative, prospective cohort study. Biol Psychiatry 2022;91:637‐46. [DOI] [PubMed] [Google Scholar]
- 232. Waszczuk MA, Docherty AR, Shabalin AA et al. Polygenic prediction of PTSD trajectories in 9/11 responders. Psychol Med 2020; doi: 10.1017/S0033291720003839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Neria Y. Functional neuroimaging in PTSD: from discovery of underlying mechanisms to addressing diagnostic heterogeneity. Am J Psychiatry 2021;178:128‐35. [DOI] [PubMed] [Google Scholar]
- 234. Bromis K, Calem M, Reinders A et al. Meta‐analysis of 89 structural MRI studies in posttraumatic stress disorder and comparison with major depressive disorder. Am J Psychiatry 2018;175:989‐98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Siehl S, King JA, Burgess N et al. Structural white matter changes in adults and children with posttraumatic stress disorder: a systematic review and meta‐analysis. Neuroimage Clin 2018;19:581‐98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Wang X, Xie H, Chen T et al. Cortical volume abnormalities in posttraumatic stress disorder: an ENIGMA‐psychiatric genomics consortium PTSD workgroup mega‐analysis. Mol Psychiatry 2021;26:4331‐43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Logue MW, van Rooij SJH, Dennis EL et al. Smaller hippocampal volume in posttraumatic stress disorder: a multisite ENIGMA‐PGC study: subcortical volumetry results from posttraumatic stress disorder consortia. Biol Psychiatry 2018;83:244‐53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Dennis EL, Disner SG, Fani N et al. Altered white matter microstructural organization in posttraumatic stress disorder across 3047 adults: results from the PGC‐ENIGMA PTSD consortium. Mol Psychiatry 2021;26:4315‐30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Kaufmann T, van der Meer D, Doan NT et al. Common brain disorders are associated with heritable patterns of apparent aging of the brain. Nat Neurosci 2019;22:1617‐23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Campbell AA, Wisco BE, Silvia PJ et al. Resting respiratory sinus arrhythmia and posttraumatic stress disorder: a meta‐analysis. Biol Psychol 2019;144:125‐35. [DOI] [PubMed] [Google Scholar]
- 241. Schneider M, Schwerdtfeger A. Autonomic dysfunction in posttraumatic stress disorder indexed by heart rate variability: a meta‐analysis. Psychol Med 2020;50:1937‐48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. McTeague LM, Lang PJ. The anxiety spectrum and the reflex physiology of defense: from circumscribed fear to broad distress. Depress Anxiety 2012;29:264‐81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243. Chen J, Zang Z, Braun U et al. Association of a reproducible epigenetic risk profile for schizophrenia with brain methylation and function. JAMA Psychiatry 2020;77:628‐36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Hannon E, Lunnon K, Schalkwyk L et al. Interindividual methylomic variation across blood, cortex, and cerebellum: implications for epigenetic studies of neurological and neuropsychiatric phenotypes. Epigenetics 2015;10:1024‐32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Iturria‐Medina Y, Khan AF, Adewale Q et al. Blood and brain gene expression trajectories mirror neuropathology and clinical deterioration in neurodegeneration. Brain 2020;143:661‐73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Tylee DS, Kawaguchi DM, Glatt SJ. On the outside, looking in: a review and evaluation of the comparability of blood and brain “‐omes”. Am J Med Genet B Neuropsychiatr Genet 2013;162B:595‐603. [DOI] [PubMed] [Google Scholar]
- 247. Morris MC, Hellman N, Abelson JL et al. Cortisol, heart rate, and blood pressure as early markers of PTSD risk: a systematic review and meta‐analysis. Clin Psychol Rev 2016;49:79‐91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Peruzzolo TL, Pinto JV, Roza TH et al. Inflammatory and oxidative stress markers in post‐traumatic stress disorder: a systematic review and meta‐analysis. Mol Psychiatry 2022;27:3150‐63. [DOI] [PubMed] [Google Scholar]
- 249. Smith AK, Ratanatharathorn A, Maihofer AX et al. Epigenome‐wide meta‐analysis of PTSD across 10 military and civilian cohorts identifies methylation changes in AHRR. Nat Commun 2020;11:5965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Katrinli S, Maihofer AX, Wani AH et al. Epigenome‐wide meta‐analysis of PTSD symptom severity in three military cohorts implicates DNA methylation changes in genes involved in immune system and oxidative stress. Mol Psychiatry 2022;27:1720‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Garrett ME, Qin XJ, Mehta D et al. Gene expression analysis in three posttraumatic stress disorder cohorts implicates inflammation and innate immunity pathways and uncovers shared genetic risk with major depressive disorder. Fron Neurosci 2021;15:678548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Breen MS, Tylee DS, Maihofer AX et al. PTSD blood transcriptome mega‐analysis: shared inflammatory pathways across biological sex and modes of trauma. Neuropsychopharmacology 2018;43:469‐81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Kuan PF, Waszczuk MA, Kotov R et al. Gene expression associated with PTSD in World Trade Center responders: an RNA sequencing study. Transl Psychiatry 2017;7:1297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Kuan PF, Clouston S, Yang X et al. Molecular linkage between post‐traumatic stress disorder and cognitive impairment: a targeted proteomics study of World Trade Center responders. Transl Psychiatry 2020;10:269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Murray GK, Lin T, Austin J et al. Could polygenic risk scores be useful in psychiatry?: a review. JAMA Psychiatry 2021;78:210‐9. [DOI] [PubMed] [Google Scholar]
- 256. Schildkraut JJ, Orsulak PJ, Schatzberg AF et al. Toward a biochemical classification of depressive disorders. I. Differences in urinary excretion of MHPG and other catecholamine metabolites in clinically defined subtypes of depressions. Arch Gen Psychiatry 1978;35:1427‐33. [DOI] [PubMed] [Google Scholar]
- 257. Schatzberg AF, Samson JA, Bloomingdale KL et al. Toward a biochemical classification of depressive disorders. X. Urinary catecholamines, their metabolites, and D‐type scores in subgroups of depressive disorders. Arch Gen Psychiatry 1989;46:260‐8. [DOI] [PubMed] [Google Scholar]
- 258. Graux P, Lorrain A, Caron J. Diagnostic and therapeutic importance of gaseous encephalography in the course of melancholic states. Echo Med Nord 1952;23:300‐1. [PubMed] [Google Scholar]
- 259. Kempton MJ, Salvador Z, Munafò MR et al. Structural neuroimaging studies in major depressive disorder. Meta‐analysis and comparison with bipolar disorder. Arch Gen Psychiatry 2011;68:675‐90. [DOI] [PubMed] [Google Scholar]
- 260. Fernandez‐Pujals AM, Adams MJ, Thomson P et al. Epidemiology and heritability of major depressive disorder, stratified by age of onset, sex, and illness course in Generation Scotland: Scottish Family Health Study (GS:SFHS). PLoS One 2015;10:e0142197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Smoller JW, Finn CT. Family, twin, and adoption studies of bipolar disorder. Am J Med Genet C Semin Med Genet 2003;123C:48‐58. [DOI] [PubMed] [Google Scholar]
- 262. Howard DM, Adams MJ, Shirali M et al. Genome‐wide association study of depression phenotypes in UK Biobank identifies variants in excitatory synaptic pathways. Nat Commun 2018;9:1470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Leung E, Lau EW, Liang A et al. Alterations in brain synaptic proteins and mRNAs in mood disorders: a systematic review and meta‐analysis of postmortem brain studies. Mol Psychiatry 2022;27:1362‐72. [DOI] [PubMed] [Google Scholar]
- 264. Howard DM, Adams MJ, Clarke TK et al. Genome‐wide meta‐analysis of depression identifies 102 independent variants and highlights the importance of the prefrontal brain regions. Nat Neurosci 2019;22:343‐52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265. Levey DF, Stein MB, Wendt FR et al. Bi‐ancestral depression GWAS in the Million Veteran Program and meta‐analysis in >1.2 million individuals highlight new therapeutic directions. Nat Neurosci 2021;24:954‐63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Labonté B, Engmann O, Purushothaman I et al. Sex‐specific transcriptional signatures in human depression. Nat Med 2017;23:1102‐11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267. Touchant M, Labonté B. Sex‐specific brain transcriptional signatures in human MDD and their correlates in mouse models of depression. Front Behav Neurosci 2022;16:845491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. Mullins N, Forstner AJ, O'Connell KS et al. Genome‐wide association study of more than 40,000 bipolar disorder cases provides new insights into the underlying biology. Nat Genet 2021;53:817‐29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269. Gordovez FJA, McMahon FJ. The genetics of bipolar disorder. Mol Psychiatry 2020;25:544‐59. [DOI] [PubMed] [Google Scholar]
- 270. Schmaal L, Pozzi E, Ho TC et al. ENIGMA MDD: seven years of global neuroimaging studies of major depression through worldwide data sharing. Transl Psychiatry 2020;10:172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Holmes SE, Abdallah C, Esterlis I. Imaging synaptic density in depression. Neuropsychopharmacology 2023;48:186‐90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272. Etkin A. A reckoning and research agenda for neuroimaging in psychiatry. Am J Psychiatry 2019;176:507‐11. [DOI] [PubMed] [Google Scholar]
- 273. Frangou S. Functional neuroimaging in mood disorders. Psychiatry 2008;8:3.18179713 [Google Scholar]
- 274. Kantrowitz JT, Dong Z, Milak MS et al. Ventromedial prefrontal cortex/anterior cingulate cortex Glx, glutamate, and GABA levels in medication‐free major depressive disorder. Transl Psychiatry 2021;11:419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275. Moriguchi S, Takamiya A, Noda Y et al. Glutamatergic neurometabolite levels in major depressive disorder: a systematic review and meta‐analysis of proton magnetic resonance spectroscopy studies. Mol Psychiatry 2019;24:952‐64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Godfrey KEM, Gardner AC, Kwon S et al. Differences in excitatory and inhibitory neurotransmitter levels between depressed patients and healthy controls: a systematic review and meta‐analysis. J Psychiatr Res 2018;105:33‐44. [DOI] [PubMed] [Google Scholar]
- 277. Truong V, Cheng PZ, Lee HC et al. Occipital gamma‐aminobutyric acid and glutamate‐glutamine alterations in major depressive disorder: an MRS study and meta‐analysis. Psychiatry Res Neuroimaging 2021;308:111238. [DOI] [PubMed] [Google Scholar]
- 278. Reddy‐Thootkur M, Kraguljac NV, Lahti AC. The role of glutamate and GABA in cognitive dysfunction in schizophrenia and mood disorders – A systematic review of magnetic resonance spectroscopy studies. Schizophr Res 2022;249:74‐84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279. Nakahara T, Tsugawa S, Noda Y et al. Glutamatergic and GABAergic metabolite levels in schizophrenia‐spectrum disorders: a meta‐analysis of 1H‐magnetic resonance spectroscopy studies. Mol Psychiatry 2022;27:744‐57. [DOI] [PubMed] [Google Scholar]
- 280. Schür RR, Draisma LW, Wijnen JP et al. Brain GABA levels across psychiatric disorders: a systematic literature review and meta‐analysis of (1) H‐MRS studies. Hum Brain Mapp 2016;37:3337‐52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Mandal PK, Guha Roy R, Samkaria A et al. In vivo 13C magnetic resonance spectroscopy for assessing brain biochemistry in health and disease. Neurochem Res 2022;47:1183‐201. [DOI] [PubMed] [Google Scholar]
- 282. Abdallah CG, Jiang L, De Feyter HM et al. Glutamate metabolism in major depressive disorder. Am J Psychiatry 2014;171:1320‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Gawryluk JW, Wang JF, Andreazza AC et al. Decreased levels of glutathione, the major brain antioxidant, in post‐mortem prefrontal cortex from patients with psychiatric disorders. Int J Neuropsychopharmacol 2011;14:123‐30. [DOI] [PubMed] [Google Scholar]
- 284. Hermens DF, Hatton SN, Lee RSC et al. In vivo imaging of oxidative stress and fronto‐limbic white matter integrity in young adults with mood disorders. Eur Arch Psychiatry Clin Neurosci 2018;268:145‐56. [DOI] [PubMed] [Google Scholar]
- 285. Lapidus KA, Gabbay V, Mao X et al. In vivo (1)H MRS study of potential associations between glutathione, oxidative stress and anhedonia in major depressive disorder. Neurosci Lett 2014;569:74‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286. Poletti S, Paolini M, Mazza MG et al. Lower levels of glutathione in the anterior cingulate cortex associate with depressive symptoms and white matter hyperintensities in COVID‐19 survivors. Eur Neuropsychopharmacol 2022;61:71‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Gigase FAJ, Snijders G, Boks MP et al. Neurons and glial cells in bipolar disorder: a systematic review of postmortem brain studies of cell number and size. Neurosci Biobehav Rev 2019;103:150‐62. [DOI] [PubMed] [Google Scholar]
- 288. Sanacora G, Yan Z, Popoli M. The stressed synapse 2.0: pathophysiological mechanisms in stress‐related neuropsychiatric disorders. Nat Rev Neurosci 2022;23:86‐103. [DOI] [PubMed] [Google Scholar]
- 289. Pu J, Liu Y, Zhang H et al. An integrated meta‐analysis of peripheral blood metabolites and biological functions in major depressive disorder. Mol Psychiatry 2021;26:4265‐76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290. Stetler C, Miller GE. Depression and hypothalamic‐pituitary‐adrenal activation: a quantitative summary of four decades of research. Psychosom Med 2011;73:114‐26. [DOI] [PubMed] [Google Scholar]
- 291. Dwyer JB, Aftab A, Radhakrishnan R et al. Hormonal treatments for major depressive disorder: state of the art. Am J Psychiatry 2020;177:686‐705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292. Sen S, Duman R, Sanacora G. Serum brain‐derived neurotrophic factor, depression, and antidepressant medications: meta‐analyses and implications. Biol Psychiatry 2008;64:527‐32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293. Brunoni AR, Lopes M, Fregni F. A systematic review and meta‐analysis of clinical studies on major depression and BDNF levels: implications for the role of neuroplasticity in depression. Int J Neuropsychopharmacol 2008;11:1169‐80. [DOI] [PubMed] [Google Scholar]
- 294. Hing B, Sathyaputri L, Potash JB. A comprehensive review of genetic and epigenetic mechanisms that regulate BDNF expression and function with relevance to major depressive disorder. Am J Med Genet B Neuropsychiatr Genet 2018;177:143‐67. [DOI] [PubMed] [Google Scholar]
- 295. Osimo EF, Pillinger T, Rodriguez IM et al. Inflammatory markers in depression: a meta‐analysis of mean differences and variability in 5,166 patients and 5,083 controls. Brain Behav Immun 2020;87:901‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296. Scott MR, McClung CA. Circadian rhythms in mood disorders. Adv Exp Med Biol 2021;1344:153‐68. [DOI] [PubMed] [Google Scholar]
- 297. Walker WH 2nd, Walton JC, DeVries AC et al. Circadian rhythm disruption and mental health. Transl Psychiatry 2020;10:28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298. Germain A, Kupfer DJ. Circadian rhythm disturbances in depression. Hum Psychopharmacol 2008;23:571‐85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299. Quera Salva MA, Hartley S. Mood disorders, circadian rhythms, melatonin and melatonin agonists. J Cent Nerv Syst Dis 2012;4:15‐26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300. McClung CA. Circadian genes, rhythms and the biology of mood disorders. Pharmacol Ther 2007;114:222‐32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301. Grandin LD, Alloy LB, Abramson LY. The social zeitgeber theory, circadian rhythms, and mood disorders: review and evaluation. Clin Psychol Rev 2006;26:679‐94. [DOI] [PubMed] [Google Scholar]
- 302. Bechtel W. Circadian rhythms and mood disorders: are the phenomena and mechanisms causally related? Front Psychiatry 2015;6:118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303. Wirz‐Justice A. Biological rhythm disturbances in mood disorders. Int Clin Psychopharmacol 2006;21(Suppl. 1):S11‐5. [DOI] [PubMed] [Google Scholar]
- 304. Lewy AJ. Circadian rhythms and mood disorders: a guide for the perplexed. J Clin Psychiatry 2015;76:e662‐4. [DOI] [PubMed] [Google Scholar]
- 305. American Psychiatric Association . Diagnostic and statistical manual of mental disorders, 5th ed. Arlington: American Psychiatric Publishing, 2013. [Google Scholar]
- 306. Nutt D, Wilson S, Paterson L. Sleep disorders as core symptoms of depression. Dialogues Clin Neurosci 2008;10:329‐36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307. Riemann D, Voderholzer U. Primary insomnia: a risk factor to develop depression? J Affect Disord 2003;76:255‐9. [DOI] [PubMed] [Google Scholar]
- 308. Mendlewicz J. Sleep disturbances: core symptoms of major depressive disorder rather than associated or comorbid disorders. World J Biol Psychiatry 2009;10:269‐75. [DOI] [PubMed] [Google Scholar]
- 309. Robillard R, Naismith SL, Smith KL et al. Sleep‐wake cycle in young and older persons with a lifetime history of mood disorders. PLoS One 2014;9:e87763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310. Maglione JE, Ancoli‐Israel S, Peters KW et al. Depressive symptoms and subjective and objective sleep in community‐dwelling older women. J Am Geriatr Soc 2012;60:635‐43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311. Tazawa Y, Wada M, Mitsukura Y et al. Actigraphy for evaluation of mood disorders: a systematic review and meta‐analysis. J Affect Disord 2019;253:257‐69. [DOI] [PubMed] [Google Scholar]
- 312. Hori H, Koga N, Hidese S et al. 24 h activity rhythm and sleep in depressed outpatients. J Psychiatr Res 2016;77:27‐34. [DOI] [PubMed] [Google Scholar]
- 313. Smagula SF, Ancoli‐Israel S, Blackwell T et al. Circadian rest–activity rhythms predict future increases in depressive symptoms among community‐dwelling older men. Am J Geriatr Psychiatry 2015;23:495‐505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314. Baek JH, Kim JS, Kim MJ et al. Lifetime characteristics of evening‐preference and irregular bed‐rise time are associated with lifetime seasonal variation of mood and behavior: comparison between individuals with bipolar disorder and healthy controls. Behav Sleep Med 2016;14:155‐68. [DOI] [PubMed] [Google Scholar]
- 315. Krane‐Gartiser K, Henriksen TE, Morken G et al. Actigraphic assessment of motor activity in acutely admitted inpatients with bipolar disorder. PLoS One 2014;9:e89574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316. Meyer N, Faulkner SM, McCutcheon RA et al. Sleep and circadian rhythm disturbance in remitted schizophrenia and bipolar disorder: a systematic review and meta‐analysis. Schizophr Bull 2020;46:1126‐43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317. Takaesu Y. Circadian rhythm in bipolar disorder: a review of the literature. Psychiatry Clin Neurosci 2018;72:673‐82. [DOI] [PubMed] [Google Scholar]
- 318. Emens J, Lewy A, Kinzie JM et al. Circadian misalignment in major depressive disorder. Psychiatry Res 2009;168:259‐61. [DOI] [PubMed] [Google Scholar]
- 319. Hasler BP, Buysse DJ, Kupfer DJ et al. Phase relationships between core body temperature, melatonin, and sleep are associated with depression severity: further evidence for circadian misalignment in non‐seasonal depression. Psychiatry Res 2010;178:205‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320. Moon J‐H, Cho C‐H, Son GH et al. Advanced circadian phase in mania and delayed circadian phase in mixed mania and depression returned to normal after treatment of bipolar disorder. EBioMedicine 2016;11:285‐95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 321. Ritter P, Soltmann B, Sauer C et al. Supersensitivity of patients with bipolar I disorder to light‐induced phase delay by narrow bandwidth blue light. Biol Psychiatry Global Open Sci 2022;2:28‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 322. Salvatore P, Ghidini S, Zita G et al. Circadian activity rhythm abnormalities in ill and recovered bipolar I disorder patients. Bipolar Disord 2008;10:256‐65. [DOI] [PubMed] [Google Scholar]
- 323. Lewy AJ, Lefler BJ, Emens JS et al. The circadian basis of winter depression. Proc Natl Acad Sci USA 2006;103:7414‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324. Lewy AJ, Rough JN, Songer JB et al. The phase shift hypothesis for the circadian component of winter depression. Dialogues Clin Neurosci 2007;9:291‐300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325. Carvalho AF, Solmi M, Sanches M et al. Evidence‐based umbrella review of 162 peripheral biomarkers for major mental disorders. Transl Psychiatry 2020;10:152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326. Serretti A, Benedetti F, Mandelli L et al. Genetic dissection of psychopathological symptoms: insomnia in mood disorders and CLOCK gene polymorphism. Am J Med Genet B Neuropsychiatr Genet 2003;121B:35‐8. [DOI] [PubMed] [Google Scholar]
- 327. Nováková M, Praško J, Látalová K et al. The circadian system of patients with bipolar disorder differs in episodes of mania and depression. Bipolar Disord 2015;17:303‐14. [DOI] [PubMed] [Google Scholar]
- 328. Maj M, Stein DJ, Parker G et al. The clinical characterization of the adult patient with depression aimed at personalization of management. World Psychiatry 2020;19:269‐93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 329. McIntyre RS, Alda M, Baldessarini RJ et al. The clinical characterization of the adult patient with bipolar disorder aimed at personalization of management. World Psychiatry 2022;21:364‐87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330. Palmer DM. Biomarkers for antidepressant selection: iSPOT‐D study. Curr Behav Neurosci Rep 2015;2:137‐45. [Google Scholar]
- 331. Dunlop BW, Binder EB, Cubellis JF et al. Predictors of remission in depression to individual and combined treatments (PReDICT): study protocol for a randomized controlled trial. Trials 2012;13:106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 332. Uher R, Huezo‐Diaz P, Perroud N et al. Genetic predictors of response to antidepressants in the GENDEP project. Pharmacogenomics J 2009;9:225‐33. [DOI] [PubMed] [Google Scholar]
- 333. Arns M, Gordon E, Boutros NN. EEG abnormalities are associated with poorer depressive symptom outcomes with escitalopram and venlafaxine‐XR, but not sertraline: results from the multicenter randomized iSPOT‐D Study. Clin EEG Neurosci 2017;48:33‐40. [DOI] [PubMed] [Google Scholar]
- 334. O'Connell CP, Goldstein‐Piekarski AN, Nemeroff CB et al. Antidepressant outcomes predicted by genetic variation in corticotropin‐releasing hormone binding protein. Am J Psychiatry 2018;175:251‐61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 335. Schatzberg AF, DeBattista C, Lazzeroni LC et al. ABCB1 genetic effects on antidepressant outcomes: a report from the iSPOT‐D trial. Am J Psychiatry 2015;172:751‐9. [DOI] [PubMed] [Google Scholar]
- 336. Maller JJ, Broadhouse K, Rush AJ et al. Increased hippocampal tail volume predicts depression status and remission to anti‐depressant medications in major depression. Mol Psychiatry 2018;23:1737‐44. [DOI] [PubMed] [Google Scholar]
- 337. Gyurak A, Patenaude B, Korgaonkar MS et al. Frontoparietal activation during response inhibition predicts remission to antidepressants in patients with major depression. Biol Psychiatry 2016;79:274‐81. [DOI] [PubMed] [Google Scholar]
- 338. Williams LM, Debattista C, Duchemin AM et al. Childhood trauma predicts antidepressant response in adults with major depression: data from the randomized international study to predict optimized treatment for depression. Transl Psychiatry 2016;6:e799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 339. Tozzi L, Goldstein‐Piekarski AN, Korgaonkar MS et al. Connectivity of the cognitive control network during response inhibition as a predictive and response biomarker in major depression: evidence from a randomized clinical trial. Biol Psychiatry 2020;87:462‐72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 340. Fischer AS, Holt‐Gosselin B, Fleming SL et al. Intrinsic reward circuit connectivity profiles underlying symptom and quality of life outcomes following antidepressant medication: a report from the iSPOT‐D trial. Neuropsychopharmacology 2021;46:809‐19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 341. Cattaneo A, Gennarelli M, Uher R et al. Candidate genes expression profile associated with antidepressants response in the GENDEP study: differentiating between baseline ‘predictors’ and longitudinal ‘targets’. Neuropsychopharmacology 2013;38:377‐85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 342. Webb CA, Dillon DG, Pechtel P et al. Neural correlates of three promising endophenotypes of depression: evidence from the EMBARC study. Neuropsychopharmacology 2016;41:454‐63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 343. Pizzagalli DA, Webb CA, Dillon DG et al. Pretreatment rostral anterior cingulate cortex theta activity in relation to symptom improvement in depression: a randomized clinical trial. JAMA Psychiatry 2018;75:547‐54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 344. Whitton AE, Webb CA, Dillon DG et al. Pretreatment rostral anterior cingulate cortex connectivity with salience network predicts depression recovery: findings from the EMBARC randomized clinical trial. Biol Psychiatry 2019;85:872‐80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 345. Nguyen KP, Chin Fatt C, Treacher A et al. Patterns of pretreatment reward task brain activation predict individual antidepressant response: key results from the EMBARC randomized clinical trial. Biol Psychiatry 2022;91:550‐60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 346. Jha MK, Minhajuddin A, Gadad BS et al. Can C‐reactive protein inform antidepressant medication selection in depressed outpatients? Findings from the CO‐MED trial. Psychoneuroendocrinology 2017;78:105‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 347. Jha MK, Minhajuddin A, Chin‐Fatt C et al. Sex differences in the association of baseline C‐reactive protein (CRP) and acute‐phase treatment outcomes in major depressive disorder: findings from the EMBARC study. J Psychiatr Res 2019;113:165‐71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 348. Grof P, Duffy A, Cavazzoni P et al. Is response to prophylactic lithium a familial trait? J Clin Psychiatry 2002;63:942‐7. [DOI] [PubMed] [Google Scholar]
- 349. Hou L, Heilbronner U, Degenhardt F et al. Genetic variants associated with response to lithium treatment in bipolar disorder: a genome‐wide association study. Lancet 2016;387:1085‐93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 350. Stone W, Nunes A, Akiyama K et al. Prediction of lithium response using genomic data. Sci Rep 2022;11:1155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 351. Teixeira AL, Colpo GD, Fries GR et al. Biomarkers for bipolar disorder: current status and challenges ahead. Expert Rev Neurother 2019;19:67‐81. [DOI] [PubMed] [Google Scholar]
- 352. Sagar R, Pattanayak RD. Potential biomarkers for bipolar disorder: where do we stand? Indian J Med Res 2017;145:7‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 353. Phillips ML, Kupfer DJ. Bipolar disorder diagnosis: challenges and future directions. Lancet 2013;381:1663‐71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 354. Anthony JC, Petronis KR. Early‐onset drug use and risk of later drug problems. Drug Alcohol Depend 1995;40:9‐15. [DOI] [PubMed] [Google Scholar]
- 355. Lopez‐Quintero C, De Los Pérez Cobos J, Hasin DS et al. Probability and predictors of transition from first use to dependence on nicotine, alcohol, cannabis, and cocaine: results of the National Epidemiologic Survey on Alcohol and Related Conditions (NESARC). Drug Alcohol Depend 2011;115:120‐30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 356. Epskamp S, van der Maas HLJ, Peterson RE et al. Intermediate stable states in substance use. Addict Behav 2022;129:107252. [DOI] [PubMed] [Google Scholar]
- 357. Lipari RN, Park‐Lee E, Van Horn S. America's need for and receipt of substance use treatment in 2015. The CBHSQ Report. Rockville: US Center for Behavioral Health Statistics and Quality, Substance Abuse and Mental Health Services Administration, 2016. [PubMed] [Google Scholar]
- 358. Moeller SJ, Platt JM, Wu M et al. Perception of treatment need among adults with substance use disorders: longitudinal data from a representative sample of adults in the United States. Drug Alcohol Depend 2020;209:107895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 359. Volkow ND. Stigma and the toll of addiction. N Engl J Med 2020;382:1289‐90. [DOI] [PubMed] [Google Scholar]
- 360. Maracic CE, Moeller SJ. Neural and behavioral correlates of impaired insight and self‐awareness in substance use disorder. Curr Behav Neurosci Rep 2021;8:113‐23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 361. Goldstein RZ, Craig AD, Bechara A et al. The neurocircuitry of impaired insight in drug addiction. Trends Cogn Sci 2009;13:372‐80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 362. Volkow ND, Koob G, Baler R. Biomarkers in substance use disorders. ACS Chem Neurosci 2015;6:522‐5. [DOI] [PubMed] [Google Scholar]
- 363. Moeller SJ, Paulus MP. Toward biomarkers of the addicted human brain: using neuroimaging to predict relapse and sustained abstinence in substance use disorder. Prog Neuropsychopharmacol Biol Psychiatry 2018;80(Pt. B):143‐54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 364. Goldstein RZ, Volkow ND. Drug addiction and its underlying neurobiological basis: neuroimaging evidence for the involvement of the frontal cortex. Am J Psychiatry 2002;159:1642‐52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 365. Wang KH, Penmatsa A, Gouaux E. Neurotransmitter and psychostimulant recognition by the dopamine transporter. Nature 2015;521:322‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 366. Freyberg Z, Sonders MS, Aguilar JI et al. Mechanisms of amphetamine action illuminated through optical monitoring of dopamine synaptic vesicles in Drosophila brain. Nature Commun 2016;7:10652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 367. Trifilieff P, Ducrocq F, van der Veldt S et al. Blunted dopamine transmission in addiction: potential mechanisms and implications for behavior. Semin Nucl Med 2017;47:64‐74. [DOI] [PubMed] [Google Scholar]
- 368. Ashok AH, Mizuno Y, Volkow ND et al. Association of stimulant use with dopaminergic alterations in users of cocaine, amphetamine, or methamphetamine: a systematic review and meta‐analysis. JAMA Psychiatry 2017;74:511‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 369. Yip SW, Kiluk B, Scheinost D. Toward addiction prediction: an overview of cross‐validated predictive modeling findings and considerations for future neuroimaging research. Biol Psychiatry Cogn Neurosci Neuroimaging 2020;5:748‐58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 370. Eisenberg IW, Bissett P, Enkavi AZ et al. Uncovering mental structure through data‐driven ontology discovery. PsyArXiv 2018; doi: 10.31234/osf.io/fvqej. [DOI] [Google Scholar]
- 371. Marek S, Tervo‐Clemmens B, Calabro FJ et al. Reproducible brain‐wide association studies require thousands of individuals. Nature 2022;603:654‐60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 372. Ivanov I, Parvaz MA, Velthorst E et al. Substance use initiation, particularly alcohol, in drug‐naive adolescents: possible predictors and consequences from a large cohort naturalistic study. J Am Acad Child Adolesc Psychiatry 2021;60:623‐36. [DOI] [PubMed] [Google Scholar]
- 373. Filippi I, Galinowski A, Lemaitre H et al. Neuroimaging evidence for structural correlates in adolescents resilient to polysubstance use: a five‐year follow‐up study. Eur Neuropsychopharmacol 2021;49:11‐22. [DOI] [PubMed] [Google Scholar]
- 374. Lees B, Garcia AM, Debenham J et al. Promising vulnerability markers of substance use and misuse: a review of human neurobehavioral studies. Neuropharmacology 2021;187:108500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 375. Blair MA, Stewart JL, May AC et al. Blunted frontostriatal blood oxygen level‐dependent signals predict stimulant and marijuana use. Biol Psychiatry Cogn Neurosci Neuroimaging 2018;3:947‐58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 376. Gonçalves SF, Turpyn CC, Niehaus CE et al. Neural activation to loss and reward among alcohol naive adolescents who later initiate alcohol use. Dev Cogn Neurosci 2021;50:100978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 377. Goldfarb EV, Scheinost D, Fogelman N et al. High‐risk drinkers engage distinct stress‐predictive brain networks. Biol Psychiatry Cogn Neurosci Neuroimaging 2022;7:805‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 378. Verdejo‐Garcia A, Albein‐Urios N. Impulsivity traits and neurocognitive mechanisms conferring vulnerability to substance use disorders. Neuropharmacology 2021;183:108402. [DOI] [PubMed] [Google Scholar]
- 379. Luciana M. Risks versus consequences of adolescent and young adult substance use: a focus on executive control. Curr Addict Rep 2020;7:453‐63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 380. Weigard AS, Brislin SJ, Cope LM et al. Evidence accumulation and associated error‐related brain activity as computationally‐informed prospective predictors of substance use in emerging adulthood. Psychopharmacology 2021;238:2629‐44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 381. Paulus MP, Tapert SF, Schuckit MA. Neural activation patterns of methamphetamine‐dependent subjects during decision making predict relapse. Arch Gen Psychiatry 2005;62:761‐8. [DOI] [PubMed] [Google Scholar]
- 382. Moeller SJ, Tomasi D, Woicik PA et al. Enhanced midbrain response at 6‐month follow‐up in cocaine addiction, association with reduced drug‐related choice. Addict Biol 2012;17:1013‐25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 383. Yip SW, Scheinost D, Potenza MN et al. Connectome‐based prediction of cocaine abstinence. Am J Psychiatry 2019;176:156‐64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 384. Carroll KM, Kiluk BD, Nich C et al. Toward empirical identification of a clinically meaningful indicator of treatment outcome: features of candidate indicators and evaluation of sensitivity to treatment effects and relationship to one year follow up cocaine use outcomes. Drug Alcohol Depend 2014;137:3‐19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 385. Reinhard I, Lemenager T, Fauth‐Buhler M et al. A comparison of region‐of‐interest measures for extracting whole brain data using survival analysis in alcoholism as an example. J Neurosci Methods 2015;242:58‐64. [DOI] [PubMed] [Google Scholar]
- 386. Parvaz MA, Moeller SJ, Malaker P et al. Abstinence reverses EEG‐indexed attention bias between drug‐related and pleasant stimuli in cocaine‐addicted individuals. J Psychiatry Neurosci 2017;42:78‐86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 387. Parvaz MA, Moeller SJ, Goldstein RZ. Incubation of cue‐induced craving in adults addicted to cocaine measured by electroencephalography. JAMA Psychiatry 2016;73:1127‐34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 388. Grimm JW, Hope BT, Wise RA et al. Incubation of cocaine craving after withdrawal. Nature 2001;412:141‐2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 389. Pickens CL, Airavaara M, Theberge F et al. Neurobiology of the incubation of drug craving. Trends Neurosci 2011;34:411‐20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 390. Versace F, Engelmann JM, Deweese MM et al. Beyond cue reactivity: non‐drug‐related motivationally relevant stimuli are necessary to understand reactivity to drug‐related cues. Nicotine Tob Res 2017;19:663‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 391. Moeller SJ, Stoops WW. Cocaine choice procedures in animals, humans, and treatment‐seekers: can we bridge the divide? Pharmacol Biochem Behav 2015;138:133‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 392. Czoty PW, Stoops WW, Rush CR. Evaluation of the “pipeline” for development of medications for cocaine use disorder: a review of translational preclinical, human laboratory, and clinical trial research. Pharmacol Rev 2016;68:533‐62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 393. Kalivas PW, Volkow ND. The neural basis of addiction: a pathology of motivation and choice. Am J Psychiatry 2005;162:1403‐13. [DOI] [PubMed] [Google Scholar]
- 394. Goldstein RZ, Volkow ND. Dysfunction of the prefrontal cortex in addiction: neuroimaging findings and clinical implications. Nat Rev Neurosci 2011;12:652‐69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 395. Garland EL, Froeliger B, Zeidan F et al. The downward spiral of chronic pain, prescription opioid misuse, and addiction: cognitive, affective, and neuropsychopharmacologic pathways. Neurosci Biobehav Rev 2013;37:2597‐607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 396. Wilcox CE, Abbott CC, Calhoun VD. Alterations in resting‐state functional connectivity in substance use disorders and treatment implications. Prog Neuropsychopharmacol Biol Psychiatry 2019;91:79‐93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 397. Moeller SJ, Bederson L, Alia‐Klein N et al. Neuroscience of inhibition for addiction medicine: from prediction of initiation to prediction of relapse. Prog Brain Res 2016;223:165‐88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 398. Wemm SE, Sinha R. Drug‐induced stress responses and addiction risk and relapse. Neurobiol Stress 2019;10:100148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 399. Blaine SK, Wemm S, Fogelman N et al. Association of prefrontal‐striatal functional pathology with alcohol abstinence days at treatment initiation and heavy drinking after treatment initiation. Am J Psychiatry 2020;177:1048‐59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 400. Parvaz MA, Rabin RA, Adams F et al. Structural and functional brain recovery in individuals with substance use disorders during abstinence: a review of longitudinal neuroimaging studies. Drug Alcohol Depend 2022;232:109319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 401. Moeller SJ, Maloney T, Parvaz MA et al. Enhanced choice for viewing cocaine pictures in cocaine addiction. Biol Psychiatry 2009;66:169‐76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 402. Moeller SJ, Maloney T, Parvaz MA et al. Impaired insight in cocaine addiction: laboratory evidence and effects on cocaine‐seeking behaviour. Brain 2010;133(Pt. 5):1484‐93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 403. Parikh A, Moeller SJ, Garland EL. Simulated opioid choice linked to opioid use disorder severity among Veterans with chronic pain: initial validation of a novel paradigm. Am J Drug Alcohol Abuse 2022;48:403‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 404. Moeller SJ, Okita K, Robertson CL et al. Low striatal dopamine D2‐type receptor availability is linked to simulated drug choice in methamphetamine users. Neuropsychopharmacology 2018;43:751‐60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 405. Balodis IM, Kober H, Worhunsky PD et al. Neurofunctional reward processing changes in cocaine dependence during recovery. Neuropsychopharmacology 2016;41:2112‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 406. Gowin JL, Harle KM, Stewart JL et al. Attenuated insular processing during risk predicts relapse in early abstinent methamphetamine‐dependent individuals. Neuropsychopharmacology 2014;39:1379‐87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 407. Kearney‐Ramos TE, Dowdle LT, Lench DH et al. Transdiagnostic effects of ventromedial prefrontal cortex transcranial magnetic stimulation on cue reactivity. Biol Psychiatry Cogn Neurosci Neuroimaging 2018;3:599‐609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 408. Kohno M, Dennis LE, McCready H et al. Dopamine dysfunction in stimulant use disorders: mechanistic comparisons and implications for treatment. Mol Psychiatry 2022;27:220‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 409. Deak JD, Johnson EC. Genetics of substance use disorders: a review. Psychol Med 2021;51:2189‐200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 410. Tsuang MT, Lyons MJ, Meyer JM et al. Co‐occurrence of abuse of different drugs in men: the role of drug‐specific and shared vulnerabilities. Arch Gen Psychiatry 1998;55:967‐72. [DOI] [PubMed] [Google Scholar]
- 411. Vollstädt‐Klein S, Wichert S, Rabinstein J et al. Initial, habitual and compulsive alcohol use is characterized by a shift of cue processing from ventral to dorsal striatum. Addiction 2010;105:1741‐9. [DOI] [PubMed] [Google Scholar]
- 412. Zhou X, Zimmermann K, Xin F et al. Cue reactivity in the ventral striatum characterizes heavy cannabis use, whereas reactivity in the dorsal striatum mediates dependent use. Biol Psychiatry Cogn Neurosci Neuroimaging 2019;4:751‐62. [DOI] [PubMed] [Google Scholar]
- 413. Lüscher C, Robbins TW, Everitt BJ. The transition to compulsion in addiction. Nat Rev Neurosci 2020;21:247‐63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 414. Gillespie NA, Neale MC, Kendler KS. Pathways to cannabis abuse: a multi‐stage model from cannabis availability, cannabis initiation and progression to abuse. Addiction 2009;104:430‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 415. Edenberg HJ, Gelernter J, Agrawal A. Genetics of alcoholism. Curr Psychiatry Rep 2019;21:26. [DOI] [PubMed] [Google Scholar]
- 416. Chand S, Gowen A, Savine M et al. A comprehensive study to delineate the role of an extracellular vesicle‐associated microRNA‐29a in chronic methamphetamine use disorder. J Extracell Vesicles 2021;10:e12177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 417. Morcuende A, Navarrete F, Nieto E et al. Inflammatory biomarkers in addictive disorders. Biomolecules 2021;11:1824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 418. Casey BJ, Cannonier T, Conley MI et al. The Adolescent Brain Cognitive Development (ABCD) study: imaging acquisition across 21 sites. Dev Cogn Neurosci 2018;32:43‐54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 419. Ostergaard S, Jensen S, Bech P. The heterogeneity of the depressive syndrome: when numbers get serious. Acta Psychiatr Scand 2011;124:495‐6. [DOI] [PubMed] [Google Scholar]
- 420. Kotov R, Cicero DC, Conway CC et al. The Hierarchical Taxonomy of Psychopathology (HiTOP) in psychiatric practice and research. Psychol Med 2022;52:1666‐78. [DOI] [PubMed] [Google Scholar]
- 421. Insel T, Cuthbert B, Garvey M et al. Research Domain Criteria (RDoC): toward a new classification framework for research on mental disorders. Am J Psychiatry 2010;167:748‐51. [DOI] [PubMed] [Google Scholar]
- 422. Botvinik‐Nezer R, Holzmeister F, Camerer CF et al. Variability in the analysis of a single neuroimaging dataset by many teams. Nature 2020;582:84‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 423. Humer E, Pieh C, Probst T. Metabolomic biomarkers in anxiety disorders. Int J Mol Sci 2020;21: 4784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 424. Breen MS, Stein DJ, Baldwin DS. Systematic review of blood transcriptome profiling in neuropsychiatric disorders: guidelines for biomarker discovery. Hum Psychopharmacol 2016;31:373‐81. [DOI] [PubMed] [Google Scholar]
- 425. Savage JE, Sawyers C, Roberson‐Nay R et al. The genetics of anxiety‐related negative valence system traits. Am J Med Genet B Neuropsychiatr Genet 2017;174:156‐77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 426. Oathes DJ, Balderston NL, Kording KP et al. Combining transcranial magnetic stimulation with functional magnetic resonance imaging for probing and modulating neural circuits relevant to affective disorders. Interdiscip Rev Cogn Sci 2021;12:e1553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 427. Xia F, Kheirbek MA. Circuit‐based biomarkers for mood and anxiety disorders. Trends Neurosci 2020;43:902‐15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 428. Adams WZ, McClure EA, Gray KM et al. Mobile devices for the remote acquisition of physiological and behavioral biomarkers in psychiatric clinical research. J Psychiatr Res 2017;85:1‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 429. Torous J, Bucci S, Bell IH et al. The growing field of digital psychiatry: current evidence and the future of apps, social media, chatbots, and virtual reality. World Psychiatry 2021;20:318‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 430. Mufford MS, van der Meer D, Andreassen OA et al. A review of systems biology research of anxiety disorders. Braz J Psychiatry 2021;43:414‐23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 431. Checkroud AM, Bondar J, Delgadillo J et al. The promise of machine learning in predicting treatment outcomes in psychiatry. World Psychiatry 2021;20:154‐70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 432. Traut N, Heuer K, Lemaître G et al. Insights from an autism imaging biomarker challenge: promises and threats to biomarker discovery. Neuroimage 2022;255:119171. [DOI] [PubMed] [Google Scholar]