Skip to main content

Abstract

Schizophrenia is a complex neuropsychiatric syndrome with a heterogeneous genetic, neurobiological, and phenotypic profile. Currently, no objective biological measures—that is, biomarkers—are available to inform diagnostic or treatment decisions. Neuroimaging is well positioned for biomarker development in schizophrenia, as it may capture phenotypic variations in molecular and cellular disease targets, or in brain circuits. These mechanistically based biomarkers may represent a direct measure of the pathophysiological underpinnings of the disease process and thus could serve as true intermediate or surrogate endpoints. Effective biomarkers could validate new treatment targets or pathways, predict response, aid in selection of patients for therapy, determine treatment regimens, and provide a rationale for personalized treatments. In this review, the authors discuss a range of mechanistically plausible neuroimaging biomarker candidates, including dopamine hyperactivity, N-methyl-d-aspartate receptor hypofunction, hippocampal hyperactivity, immune dysregulation, dysconnectivity, and cortical gray matter volume loss. They then focus on the putative neuroimaging biomarkers for disease risk, diagnosis, target engagement, and treatment response in schizophrenia. Finally, they highlight areas of unmet need and discuss strategies to advance biomarker development.
Schizophrenia is a complex neuropsychiatric syndrome with a heterogeneous genetic, neurobiological, and phenotypic profile. Currently, no objective biological measures—that is, biomarkers—are available that inform diagnostic or treatment decisions. A biomarker, as outlined by the U.S. Food and Drug Administration and National Institutes of Health Biomarker Working Group, is “a characteristic that is measured as an indicator of normal biological processes, pathogenic processes, or responses to an exposure or intervention, including therapeutic interventions” (1). Neuroimaging is a strong candidate for biomarker development in schizophrenia. Imaging can capture phenotypic variations in molecular and cellular disease targets or in brain circuits that are a unique representation of gene-environment interactions and are associated with behavioral alterations (2). It offers versatility in terms of measuring multiple pathophysiological mechanisms, including brain structural integrity deficits, functional dysconnectivity, and altered neurotransmitter systems (Figure 1) (3). For a biomarker to be practically useful, it must be a proxy of a clinically relevant measure. It needs to have an acceptable level of sensitivity, specificity, and predictive value (4). Ideally, it will also be easily quantifiable and cost-effective.
FIGURE 1. An introduction to relevant terms in neuroimaginga
a SPECT=Single-photon emission computed tomography.
In a 2012 consensus report, the APA Work Group on Neuroimaging Markers of Psychiatric Disorders suggested a number of criteria that should be met in order to establish the validity of a neuroimaging biomarker. A diagnostic biomarker should have a sensitivity >80% in detecting a particular psychiatric disorder, a specificity >80% in distinguishing this disorder from other psychiatric disorders, and a positive predictive value that approaches 90%. Furthermore, the data used to establish a biomarker should require confirmation by at least two independent sets of qualified investigators, with results published in peer-reviewed journals (5). To date, no neuroimaging biomarker has met these criteria. Nonetheless, a number of studies have made progress toward the goal of biomarker development in schizophrenia.

Mechanistically Plausible Targets for Biomarker Development in Schizophrenia

Mechanistically based biomarkers represent a direct measure of the pathophysiological underpinnings of the disease process (6) and thus can serve as true intermediate or surrogate endpoints (Figure 2). These biomarkers can validate new treatment targets or pathways, predict treatment response, aid in selection of patients for therapy, determine therapeutic regimens, and provide a rationale for personalized treatments (7). Ideally, biomarker development targets would reflect fundamental neurobiological alterations, have analogues in preclinical models, correlate with measures of clinical symptom severity, and be consistent with models of disease pathology (8). Here, we selectively review mechanistically plausible biomarker development targets that meet these criteria (Table 1).
FIGURE 2. Mechanistically plausible neuroimaging biomarker development targetsa
a Based on discovery-oriented research, a number of mechanistically plausible neuroimaging biomarker development targets have been identified. Diagnostic biomarkers aid in the detection of the presence of a disease state. Target engagement biomarkers confirm therapeutic properties of drugs and can be used for determination of appropriate dosing. Treatment response prediction biomarkers guide the appropriate choice of treatment. Therapeutic monitoring biomarkers monitor the response to a specific treatment or monitor relapse of a disorder. NMDAR=N-methyl-d-aspartate receptor.
TABLE 1. Mechanistically plausible targets for biomarker development in schizophreniaa
Putative Biomarker TargetModel of Disease PathologyPreclinical ModelAssociation With Clinical Variables
Dopamine hyperactivityDopaminergic hyperactivity in subcortical and limbic brain regions results in positive symptomsAmphetamine modelAssociations are reported between dopamine dysregulation and psychosis severity; D2 receptor occupancy predicts antipsychotic treatment response
NMDA receptor hypofunctionNMDA receptor hypofunction on GABAergic interneurons causes disinhibition of the glutamatergic pyramidal cellNMDA receptor antagonist (ketamine, PCP, MK-801) modelsAssociations are reported between glutamate levels and subsequent treatment response
Hippocampal hyperactivityA hyperglutamatergic state causes hippocampal hyperactivity, which then results in downstream dopamine circuit dysregulationMethylazoxymethanol acetate modelAssociations are reported between hippocampal regional cerebral blood flow and psychosis symptom severity as well as treatment response
Neuroinflammation/immune dysregulationMicroglia are primed to act in a hyperresponsive manner and shift to a proinflammatory state in response to stress, resulting in aberrant neurotransmission and structural injuryMaternal immune activation modelsInflammation appears more prominent in early illness stages and in patients with acute psychosis symptom exacerbations
DysconnectivityDisturbances in the excitation/inhibition balance result in altered functional network architectureNMDA receptor antagonist models, fractalkine knockout modelAssociations are reported between dysconnectivity and psychosis symptom severity as well as treatment response
Cortical gray matter lossAbnormalities in brain maturational processes mediated by environmental factors results in gray matter lossSubchronic PCP antagonism, maternal immune activation modelsAssociations are reported between gray matter loss and disease severity and are linked to overall poor outcomes
a
GABA=γ-aminobutyric acid; NMDA=N-methyl-d-aspartate; PCP=phencyclidine.
Dopamine hyperactivity has a long history as a prominent pathophysiologic hypothesis of schizophrenia (9, 10), as medications that treat psychosis are dopamine D2 receptor antagonists (11) and dopamine-enhancing agents such as stimulants are psychotomimetic (12). In rodent models, amphetamine administration induces locomotor sensitization that is accompanied by an increase in dopamine efflux from the nucleus accumbens and dorsal striatum (13). In schizophrenia patients, a link between dopamine dysregulation and psychosis severity (14) and a relationship between baseline dopamine D2 receptor occupancy and antipsychotic treatment response have been reported (1517).
N-Methyl-d-aspartate receptor (NMDAR) hypofunction is widely hypothesized to be a central neurobiological alteration in schizophrenia (18, 19). Experimental evidence supports NMDAR hypofunction as a high-priority target for biomarker development. NMDAR hypofunction on the γ-aminobutyric acid (GABAergic) interneuron causes disinhibition of the glutamatergic pyramidal cell (2022). The presence of a hyperglutamatergic state in different brain areas in patients with schizophrenia has been empirically confirmed and replicated in a number of magnetic resonance spectroscopy (MRS) studies (2329). Preclinical data further support this target by demonstrating that experimentally induced NMDAR hypofunction results in increased firing of glutamatergic neurons in animal models (30) and produces psychosis-like behavioral phenotypes (3133) and glutamatergic excess in healthy human subjects (3436). Unfortunately, no validated positron emission tomography (PET) ligand visualizing NMDAR function in vivo is available to date (37).
Another biomarker development target is hippocampal hyperactivity. Here, the model suggests that a hyperglutamatergic state causes hippocampal hyperactivity, which then may result in downstream dopamine circuit dysregulation and psychotic symptoms (38, 39). Several studies reported hippocampal hyperactivity in patients with schizophrenia (4043) and found a relationship between hippocampal regional cerebral blood flow and psychosis symptom severity (44) as well as antipsychotic treatment response (45). Linking cellular-level mechanisms and neuroimaging findings, Schobel and colleagues (46) reported a series of experiments in a preclinical model of psychosis showing that ketamine administration causes an increase in extracellular glutamate and hippocampal hyperactivity. Experiments in a methylazoxymethanol acetate model, which recapitulates a developmental disruption leading to neurophysiological and behavioral deficits that resemble components of schizophrenia, further demonstrate that hippocampal hyperactivity results in increased dopaminergic signaling, which can be reversed by inactivating the ventral hippocampus (47), suggesting possible translational utility of this marker.
Neuroinflammation or immune dysregulation as plausible biomarkers for schizophrenia are rooted in the observation of a link between autoimmune processes and development of psychosis (48). In this model, microglia are primed during early development into a hyperresponsive mode, then shift to a proinflammatory state in response to stress during critical developmental periods. This in turn can result in aberrant neurotransmission, synaptic pruning, and structural injury of neurons and glia (49). Maternal immune activation models in rodents show a postpubertal symptom onset and have structural, neurochemical, and behavioral abnormalities recapitulating the human clinical picture (50). Evidence of immune dysregulation has also been reported in postmortem studies (51) and in studies examining cytokines in cerebrospinal fluid of patients (52). A meta-analysis of PET studies using translocator protein binding, which is expressed during the inflammatory response, found significantly elevated tracer binding in patients with schizophrenia compared with control subjects, with a small to moderate effect size (Hedges’ g=0.31), but no difference in volume of distribution was detected (53), although another study (54) did not report evidence of neuroinflammation in the dorsolateral prefrontal cortex or hippocampus in unmedicated patients after controlling for relevant genetic polymorphisms. Two studies have reported evidence of neuroinflammation in psychosis spectrum patients with recent onset and patients with acute illness (55, 56). Interestingly, these alterations are not found in medicated patients with chronic illness (57, 58), suggesting a dynamic imbalance. This is further supported by a number of diffusion imaging studies reporting increased extracellular free water, a proxy of inflammation, which appears more prominent in the early illness compared with chronic stages (5962). Taken together, biomarkers related to immune dysregulation may be dynamic, in that they capture pathological processes that are present only during illness onset or psychosis exacerbations.
Human connectome studies have provided data concordant with the hypothesis that brain network dysconnection is fundamental to psychosis (6365). The dysconnectivity model proposes that NMDAR-mediated disturbances in the excitation/inhibition balance lead to altered functional network architecture, which in turn results in the symptoms of schizophrenia (66, 67). A number of studies have demonstrated a relationship between brain network dysconnectivity and the development of psychotic symptoms (62), symptom severity (68), and response to antipsychotic pharmacotherapy (6971). Lending additional support to this model are studies that report disruption in functional brain networks following experimentally induced NMDAR hypofunction in healthy human subjects (35, 72, 73). While dysconnectivity classically is explained by changes in neurotransmitter systems and aberrant modulation of synaptic efficiency (66), contemporary theories also consider the underlying anatomical connections. Prominent neurodevelopmental models postulate that genetic and environmental factors may affect early white matter developmental trajectories followed by the onset of psychosis, which then results in further reductions in white matter integrity (7477). Those perturbations result in white matter development disruptions and altered behavioral phenotypes, supporting this neurodevelopmental model (78). For example, mice lacking the chemokine receptor fractalkine exhibit a transient reduction of microglia and a deficit in synaptic pruning, which results in decreased connectivity between the frontal cortex and hippocampus and deficits in social interactions (79). Importantly, diffusion imaging studies report associations between white matter abnormalities and disease severity across symptom dimensions (8084), poor response to antipsychotic treatment (85), and worse overall outcomes (86, 87).
Similarly, cortical gray matter volume loss, which may be most prominent in fronto-temporal regions, is consistently found as a hallmark feature that is already present at illness onset and may become progressively worse with longer illness duration (88, 89). Abnormalities in brain maturational processes mediated by environmental factors are hypothesized to underlie gray matter alterations (90). Subchronic NMDAR antagonism (91, 92) and maternal immune activation models (93) have resulted in gray matter reductions in preclinical studies. Gray matter loss has been linked to disease severity across symptom dimensions (9496) and overall poor clinical outcomes (97), suggesting that it may be a viable candidate for diagnostic and prognostic biomarker development.
We have discussed a number of mechanistically plausible biomarker development targets, but it needs to be acknowledged that this is not an exhaustive list. Other targets are also plausible, such as oxidative stress or GABA dysfunction, where a disruption in the fast-spiking GABAergic interneurons has strong evidentiary support in the postmortem schizophrenia literature, and parvalbumin-positive interneuron dysfunction has been identified as a common factor behind several of the relevant animal models (98, 99). As we next discuss, the specific utility of any of these markers may depend on the phase of illness and the specific clinical question to be answered.

Biomarkers for Risk Prediction

Correct identification of individuals at risk for psychosis in the prodromal phase provides a major opportunity for early intervention and disease prevention. However, when solely relying on clinically high-risk criteria, correct disease prediction is estimated to be 15%−30% (100). In a two-center study examining structural MRI for predicting transition risk in patients at high risk for developing schizophrenia, Koutsouleris and colleagues (101) demonstrated a statistically significant improvement in prognostic certainty over clinical assessments alone. The anatomical features associated with a later transition to schizophrenia included gray matter reductions in the prefrontal, cingulate, striatal, and cerebellar cortices. However, the study’s pooled sample was unusual in the fact that approximately 45% of the enrolled subjects transitioned to a psychotic illness, which is substantially higher than in a typical sample for this type of study (101); replication in independent samples will therefore be important.
In a small study, Schobel and colleagues (102) found that cerebral blood volume in the CA1 subfield of the hippocampus predicted clinical progression with a positive predictive value of 71% and a negative predictive value of 82%. The same group later reported that left anterior CA1 cerebral blood volume also predicted time to psychosis onset in high-risk patients, and demonstrated this to be a more sensitive marker of clinical outcomes compared with subthreshold psychotic symptoms (46).
Thalamic glutamate measurements with MRS in a relatively small sample of ultra-high-risk individuals predicted the clinical course with an odds ratio of 0.52, such that higher baseline glutamate levels were associated with subsequent remission of prodromal symptoms (103). In contrast, high-risk subjects who later developed a frank psychotic episode showed higher glutamate levels in the dorsal caudate compared with the nontransition group and control subjects. That study reported a surprisingly large effect size (Cohen’s d) of 1.39 (104). Another small study showed that clinical high-risk individuals who later became psychotic had higher hippocampal glutamate levels compared with those who did not transition; the effect size (Hedges’ g) was 0.57 (105), suggesting that brain regions are an important consideration when developing spectroscopy-based biomarkers.

Diagnostic Biomarkers

The goal in developing diagnostic biomarkers is to detect the presence of a disease state and to establish objective disease signatures. A meta-analysis of multivariate pattern recognition studies of neuroimaging-based diagnostic biomarkers found that these markers separate patients from control subjects with an overall sensitivity and specificity of approximately 80%. Interestingly, the study authors reported that sensitivity was higher in patients with chronic illness compared with first-episode patients, and they found potential effects of symptom severity and antipsychotic dosage on specificity, suggesting that these biomarkers may be better in correctly classifying patients with a higher overall disease burden (106). Leveraging the Functional Biomedical Informatics Research Network Data Repository (fBIRN), Calhoun and colleagues (107) demonstrated that a multimodal classifier, which combined structural MRI and resting-state functional MRI, improved classification accuracy when compared with each modality used separately. This suggests that an aggregate approach to biomarker development may be fruitful.

Target Engagement Biomarkers

Target engagement biomarkers are meant to confirm desired therapeutic properties or to determine individualized dosing for a specific treatment. To qualify as a target engagement biomarker, it must measure a direct interaction between the treatment and the intended molecular or functional target in the central nervous system, which for medications is typically accomplished with PET. However, any neuroimaging outcome measure is theoretically an engageable target, as long as it has the potential to change measurably with a targeted intervention.
PET studies of the dopaminergic system provided the classic demonstration of target engagement for antipsychotic medications through several lines of converging evidence. These included establishment of abnormal dopaminergic function in patients with schizophrenia (17, 108), discovery of a link between dopamine dysregulation and psychosis severity (14), and demonstration of a reduction in dopamine stimulation of D2 receptors with antipsychotic medication treatment (109).
In the absence of a specific PET ligand targeting the glutamatergic system, a number of MRI markers are being used as a proxy for glutamate. The Fast-Fail Trials in Psychotic Spectrum Disorders biomarker project, based on the hypothesis of glutamatergic neurotransmitter system disturbances as a core pathological feature in schizophrenia (18), approached validation of target engagement in two phases. In the first stage, three candidate biomarkers that are putatively sensitive to glutamatergic alterations were evaluated for their power to detect changes resulting from experimentally induced NMDAR hypofunction using a ketamine challenge (110). The effect size was most robust and consistent across sites for a resting-state blood-oxygen-level-dependent (BOLD) signal response in the dorsal anterior cingulate cortex, suggesting possible utility of this measure as a biomarker for glutamatergic change. Importantly, a mean signal change of 0.5% fully separated subjects receiving ketamine from those receiving placebo. In the second stage, this marker was used to enrich the sample by eliminating subjects who did not demonstrate a ketamine-induced BOLD signal change before randomizing healthy volunteers to receive either pomaglumetad, a partial mGluR-2/3 agonist, or placebo. Here, the goal was to test the hypothesis that the drug would blunt the ketamine-induced BOLD response, indicating its glutamate target engagement, and to determine dosing of the medication. This stage of the study has now completed data collection; pending results will inform the decision for pomaglumetad to be advanced into further trials.

Biomarkers to Predict Response to Treatment or to Aid in Therapeutic Monitoring

This type of biomarker is intended to characterize patients in the context of a given treatment before it is started, and it may be used for patient stratification into biomarker positive and negative groups. In addition, these biomarkers may also be used to monitor the effectiveness of treatment or to predict deleterious side effect occurrence and probability of relapse (111).
Antipsychotic medications principally act on dopamine D2 receptors, which informed the dopamine hyperactivity hypothesis of schizophrenia. A number of studies reported a relationship between dopamine D2 receptor occupancy at baseline and subsequent clinical response to antipsychotic medications (15, 17), and a relationship between D2 receptor occupancy with antipsychotic medication treatment and the extent of clinical improvement (15, 16). Interestingly, the level of haloperidol-induced D2 receptor blockage has also been found to be associated with the degree of extrapyramidal symptom severity and prolactin elevation (15). Taken together, the data suggest that dopamine D2 receptor occupancy is both a mediator and a moderator of antipsychotic treatment response.
The schizophrenia literature links greater reductions in gray matter volume to worse long-term clinical outcomes. In first-episode psychosis patients, a machine learning classifier based on gray matter morphometry found baseline gray matter volumes in the left and right parahippocampal gyri to be most important for predicting remission status after 6 months of antipsychotic treatment, with an accuracy of 79% (112). Similarly, a multivariate morphometry study found that lower gray matter volume in the left and right inferior frontal gyrus and anterior insula predicted a lack of improvement in symptoms over 1 year (113). The gyrification index, a gray matter morphometric feature acting as a surrogate of neurodevelopmental trajectories, was found to be altered in schizophrenia (114) and possibly sensitive to antipsychotic exposure (115). In first-episode psychosis, reduced gyrification in frontal, insular, and temporal cortices has been associated with subsequent poor response to antipsychotic treatment (116), suggesting that it may be a useful predictor of treatment response.
A growing body of evidence suggests the potential to predict treatment response using glutamatergic neurometabolites measured with MRS. OPTiMiSE, a European multicenter study, found that higher baseline glutamate levels in the anterior cingulate cortex in first-episode psychosis patients were associated with greater symptom severity and lower likelihood of remission after 4 weeks of antipsychotic treatment (117). This is consistent with a previous report by the same group finding 11% higher glutamate levels in the same region in a small group of patients with medication-resistant illness compared with a group with medication-responsive illness (Cohen’s d=0.76), although accurate classification of response status was not possible with this measure (118). In a 2017 review of longitudinal spectroscopy studies, Egerton and colleagues (119) reported an overall mean reduction in brain Glx (glutamate + glutamine) of 6.5% after antipsychotic treatment and suggested that antipsychotic treatment response may be associated with lower glutamate levels before treatment and greater reductions in glutamate with treatment. However, it is possible that effects are region specific, as others have reported that in the hippocampus, higher as opposed to lower baseline Glx may be associated with favorable treatment response (25).
A promising resting-state-fMRI-based putative biomarker was recently described in a series of experiments examining functional connectivity of the striatum, a principal site of antipsychotic drug action. First, Sarpal and colleagues (120) demonstrated an increase in striatal connectivity after 16 weeks of antipsychotic treatment, and this change was correlated with the degree of clinical symptom improvement. After this, the authors created a baseline “striatal connectivity index” that predicted subsequent treatment response in first-episode psychosis patients. This index was then tested for its utility in separating good from poor treatment responders in an independent cohort of hospitalized patients with chronic schizophrenia. Notably, this index showed a significant separation between good and poor responders, with a positive predictive value of 76% and a negative predictive value of 79% (71). Extending this work, others later reported lower striatal connectivity in schizophrenia patients with antipsychotic treatment-resistant illness compared with patients with nonrefractory illness (121).
A number of resting-state fMRI studies have suggested that the connectivity and topology of several other known brain networks (69, 70, 122125) may also have utility in predicting response to antipsychotic treatment, although the performance of the majority of these putative markers remains to be established at the single-subject level. The only study to date investigating the accuracy of resting-state cortical connectivity at the single-subject level reported that baseline functional connectivity between the superior temporal cortex and other cortical regions predicted clinical response after 10 weeks of antipsychotic treatment, with a balanced accuracy of 82% (126). Another group used a non-connectivity-based resting-state index, amplitude of low-frequency fluctuations (ALFF), to build a machine learning classifier predicting treatment response in patients with recent-onset schizophrenia. The authors reported that ALFF in the left postcentral gyrus/inferior parietal lobule predicted response status with an accuracy of 72.7%. A major strength of the study was the independent replication sample (using a different scanner) that showed a similar accuracy of 75% in predicting remission status (127).

Challenges in Biomarker Development

Three challenges have limited the development of neuroimaging biomarkers in schizophrenia: internal heterogeneity, analytic approaches, and clinical utility.
First, one of the principal challenges of diagnostic biomarker research is that the standard nosology in schizophrenia is based on symptoms alone. There is an inherent tension between the effort to develop biomarkers for classic DSM-based diagnoses and the growing sense that it may be more fruitful to develop biomarkers that identify more biologically homogeneous subgroups of patients (128). In an effort to overcome these inherent limitations, the Research Domain Criteria paradigm was introduced as an alternative framework for the investigation of psychiatric disorders (129), in which disorders are considered in terms of disruptions of normal-range operation systems. Alternatively, the Bipolar-Schizophrenia Network on Intermediate Phenotypes initiative has incorporated a dimensional approach in an effort to analyze biomarker outcomes for disease definition and neuropathology in psychosis spectrum disorders (130). Findings support the idea that neuroimaging-based biomarkers do not obey the boundaries set by symptom-based nosology, underscoring the limitations of diagnostic biomarker development studies conducted within the context of traditional diagnostic boundaries.
Closely related is the complexity of the underlying pathophysiology. Because of the high degree of intricate associations between neurotransmitter systems, immune systems, functional brain network architecture, and brain structure, a biomarker may reflect the result of multiple modulatory inputs rather than the primary etiological factor (131). Unfortunately, multivariate approaches capitalizing on the vast amount of multimodal neuroimaging data available to detect representative pathway biomarkers assessing key pathological features such as NMDAR hypofunction, inflammation, or neuronal plasticity have yet to be developed. It also remains unclear whether putative biomarkers assessed in a clinically stable state or in a more actively psychotic state will be most informative. Furthermore, the cross-sectional assessment of the candidate marker may not be the most relevant because the change of a given marker over time in an individual may be most predictive. Additionally, it is possible that certain relevant biological signatures, such as alterations in spine density or synaptic integrity (132, 133), are simply below the resolution that can be captured by existing imaging methods.
The second challenge lies in defining standard best practices for analytics, both in preprocessing of the original data and in the development of prediction rules. To the first point, the majority of conventional imaging indices are affected by scanner-specific measurement errors, which can have a significant impact on cross-site reliability. Furthermore, there is a lack of consensus in the field on how to harmonize imaging sequences or mitigate measurement error in the postprocessing stage across sites, which greatly reduces biomarker accuracy and reliability in multisite studies. To the second point, many biomarker studies do not report their results in the most rigorous fashion. The ultimate goal of every biomarker is to predict an unseen future: conversion to first-episode psychosis, response to a specific medication, long-term prognosis, and so on. The most rigorous approach to assessing that prediction performance is replication of a biomarker on a new data set, collected by researchers independent of the original team. Since that type of replication generally takes years, a close alternative is cross-validation: reporting a prediction model’s performance on new data it has never seen before (as opposed to the data originally used to build the model) (134, 135). Without cross-validation, biomarker studies are susceptible to the problem of “overfitting”—building models that work very well on one specific sample but do not generalize to the larger clinical population. Unfortunately, cross-validation remains rare in schizophrenia biomarker studies, as it does in psychiatric biomarker research more generally.
The third challenge lies in determining the clinical utility of a biomarker. The decision to use a biomarker in clinical practice should be based on an expectation that it will have a positive health impact; therefore, measuring biomarker performance in generic terms is not sufficient for confirmation of clinical utility (136). Demonstration of clinical utility is typically a two-step process that first determines the accuracy of a biomarker and then shows that using the biomarker information in managing patients, given the benefits and risks associated with the assessment, improves outcomes in a clinically meaningful way (137). Prospective, confirmatory multicenter studies or decision-modeling approaches could be used to demonstrate clinical utility (136, 137), but these types of studies are clearly a missing element in the evaluation of neuroimaging biomarkers in the field.
Of course, these challenges are not just specific to neuroimaging biomarker development in schizophrenia; they largely apply to neuroimaging biomarker research in general. Similarly, the following section, where we discuss next-generation biomarker development studies and reflect on best practices, is also applicable to the broader field.

Next-Generation Biomarker Development Studies

Several lines of research are well positioned to contribute to the advancement of neuroimaging biomarker discovery (Figure 3).
FIGURE 3. Next generation of neuroimaging biomarker studies in schizophrenia
Many of the routinely assessed imaging markers are difficult to link directly to relevant pathological processes. For example, glutamate levels measured with spectroscopy do not equal the amount of glutamatergic neurotransmission, but rather reflect the amount of neuronal, glial, and synaptic glutamate present in a voxel. BOLD imaging does not measure neural activity directly. It is a signal that stems from a complex interaction between blood flow, blood volume, and oxygen consumption related to neural activity. The field has demonstrated a commitment to the long-term goal of creating the next generation of imaging tools that allow us to map the brain and relevant pathological signatures on a much more granular level (138), which will likely accelerate biomarker discovery. Continuing development of innovative technologies that are biologically validated and capture relevant features of complex neural circuits will provide unique opportunities. Here, radiotracer development holds distinct promise, as novel pharmacotherapeutics will likely target modulation of neurotransmitter systems. Development of specific ligands that capture principal pathophysiological processes such as NMDAR hypofunction or immune dysregulation could result in significant advancement of diagnostic and target-engagement biomarker research. Another strategy that may have greater short-term feasibility is to capitalize on already existing imaging techniques and leverage advances in computational modeling to increase the specificity of measures. For this strategy to be successful, vital components include testing the biological accuracy by rigorous histological validation and confirming disease relevance in preclinical models.
It will be critical to design studies that follow sound methodology to minimize bias and maximize precision (139). Unfortunately, this is not a trivial undertaking with neuroimaging data, as factors such as magnetic field strength, sequence acquisition parameters, scanner variability, and data analysis methods affect measurements. Additionally, head movement has a negative impact on data quality in virtually all neuroimaging modalities (140). A number of these problems can be mitigated by implementation of standardized image acquisition protocols, use of imaging phantoms as external reference if feasible, rigorous data quality control protocols, retrospective sequence harmonization (141), and standardized data processing pipelines and data analytics.
Future studies would benefit from including efforts to cross-validate biomarkers in independent samples not only in patients with comparable clinical characteristics but also with various clinical presentations (acutely psychotic, clinically stable), at early and late disease stages, and with differential antipsychotic medication exposure. This type of data would be invaluable in delineating the temporal evolution and scope of a candidate biomarker.
Because an aggregate approach, either as a combination of multiple imaging modalities or the addition of information from serological or genetic markers, could improve biomarker performance, an important next step beyond independent replication will be to embed a number of biomarkers in a predictive model and to test overall performance, discrimination, calibration, and clinical utility of the model (142). An excellent example of the important considerations of predictive models, in this case clinical risk prediction models for conversion to psychosis (142), could also serve as a road map for building analogous models for disease risk or therapeutic response that are neuroimaging biomarker based.
Conventional statistical approaches for three-dimensional neuroimaging data rely on mass-univariate analyses (143). Machine learning, a field in artificial intelligence, is designed to detect patterns in data with multivariate statistics aimed at making predictions at the single-subject level. To date, the majority of machine learning studies have focused on correctly classifying the presence of a disease state. However, the practical value here is limited, as those patients are already “correctly classified” through clinical assessments. Studies geared toward building models that can accurately predict the risk for disease development, clinical course, or a patient’s likelihood of responding to a given treatment may be more clinically useful but have been less common (144). There are also a number of technical limitations that constrain interpretability of machine learning studies in schizophrenia. Because overfitting of data is a major source of bias, it is important that sample sizes are adequate when developing a model and that the model is then cross-validated as noted above (145). Another limitation of machine learning approaches is that many studies use a “black box” approach, reporting sensitivity, specificity, and accuracy of an imaging classifier without providing spatial maps outlining relevant features of classifiers. Even though these types of complementary analyses are computationally expensive, they would make models more interpretable and add face validity to them. This idea of adding “explainability” to artificial intelligence techniques is a major priority for federal funders whose investments often shape a field (146). Computer science holds immense promise in providing solutions for these technical limitations in the next generation of biomarker discovery studies. Opportunities are in the optimization of informatics infrastructures or design of user-friendly software interfaces geared toward neuroscientists with limited training in command-line based coding, and in interdisciplinary collaborations guiding appropriate implementation of multivariate statistics.
The literature on best practices in how to objectively and effectively evaluate the clinical utility of a neuroimaging biomarker in schizophrenia is sparse. Defining biomarker performance standards based on established decision theory concepts has shown potential to inform clinically relevant levels of sensitivity and specificity in cancer research. We would argue against arbitrary targets in favor of calculating target levels for a given intended use of a biomarker, for example, screening versus confirmatory testing. These calculations consider variables such as prevalence of the disease, which affects the true positive and negative predictive values, and the cost-benefit ratio of such tests (147). Additionally, performance of a biomarker in one context may not be relevant to the setting of interest (148). Development of calculators for estimation of individualized minimally acceptable biomarker performance indicators and necessary sample size based on these considerations could be invaluable in the design of future biomarker studies.
Lastly, to accelerate biomarker development, it is important to address transparency and reproducibility of research findings. On a policy level, many funding agencies now mandate sharing of generated data with the scientific community. Unfortunately, the sharing process is quite laborious at this time. Refining electronic data capture systems to allow highly automated sharing and integration of data sharing and analysis tools will improve reproducibility of imaging findings (149). It will also be important to develop reporting standards containing the minimum recommendations for biomarker development studies, similar to those for randomized clinical trials (150) and systematic reviews (151), which are widely recognized in the scientific community. These guidelines help improve the quality of reporting in scientific journals by outlining essential items necessary for a clear and transparent account of research methods and results. Another interesting idea that could address reproducibility issues for future studies is the creation of a preregistration database for neuroimaging biomarker studies with the original hypothesis and its justification, similar to what is done in clinical trials (152).
Moving forward, nationwide or multinational initiatives supporting large-scale studies that are developing targeted biomarkers have real potential to individualize clinical care in this complex neuropsychiatric syndrome. In the field of Alzheimer’s disease, the Alzheimer’s Disease Neuroimaging Initiative (ADNI), a private-public partnership, was designed to develop biomarkers for the early detection and tracking of the disease. The investment of $218 million to date has enabled ADNI to discover biomarkers for use in clinical trial subject selection and as surrogate outcome measures, to develop standardized protocols for use across multiple centers, to create platforms allowing open data access, and to make advances in the understanding of the relationship between biomarkers and disease progression (153). This initiative could serve as a road map for the design of biomarker development studies for schizophrenia spectrum disorders.

Acknowledgments

The authors thank Dr. John Krystal for his thoughtful feedback on the first draft of the manuscript, and Dr. Anthony Vernon for sharing his expertise on animal models.

Footnotes

This article is derived from work done by the Work Group for Biomarkers and Novel Treatments of the APA Council of Research on behalf of the American Psychiatric Association (APA) and remains the property of the APA. It has been altered only in response to the requirements of peer review. Copyright © 2020, American Psychiatric Association. Published with permission.
The views expressed in this article are those of the authors and do not necessarily represent the views of the officers, trustees, or all members of the American Psychiatric Association.
Dr. Kraguljac has served as a consultant for Neurocrine Biosciences. Dr. McDonald has received research support from NIMH, the National Institute of Neurological Disease and Stroke (NINDS), the National Institute on Aging (NIA), the Patient-Centered Outcomes Research Institute, Cervel Neurotherapeutics, NeoSync, Neuronetics, Soterix, and the Stanley Foundation; he is an ad hoc member of several NIMH and NINDS study sections and a member of the APA Council on Research and Quality representing ECT and Neuromodulation Therapies; he serves on the boards of 3Keys and Skyland Trail; he serves as chair of the data safety monitoring board for the NIA multicenter study; he receives royalties from Oxford University Press; and he has an endowed chair funded by the J.B. Fuqua Foundation. Dr. Widge has received support from the Minnesota’s Discovery, Research, and InnoVation Economy (MnDRIVE) initiative, the Minnesota Medical Discovery Team on Addictions, and NIH; he has received device donations from Medtronic; he has served as a consultant for Circuit Therapeutics, Cyberonics, and Medtronic; and he has multiple patent applications in the area of brain stimulation and circuit modification to improve cognition. Dr. Rodriguez has served as a consultant for Epiodyne, receives research grant support from Biohaven Pharmaceuticals, and serves as a Deputy Editor at the American Journal of Psychiatry. Dr. Tohen has served as a consultant for Abbott, Alexza, Alkermes, Allergan, AstraZeneca, Bristol-Myers Squibb, Elan, Eli Lilly, Forest, Gedeon Richter, GlaxoSmithKline, Intra-Cellular Therapies, Johnson & Johnson, Lundbeck, Merck, Minerva, Otsuka, Pamlab, Roche, Sunovion, Teva, and Wyeth. Dr. Nemeroff has received grants or research support from NIH and the Stanley Medical Research Institute; he has served as a consultant for Acadia Pharmaceuticals, Aditum Bio, Axsome, BioXcel Therapeutics, Bracket (Clintara), Dainippon Pharma, EMA Wellness, Fortress Biotech, Intra-Cellular Therapies, Janssen Research and Development, Magstim, Navitor, Prismic Pharmaceuticals, Sage, Signant Health, Silo Pharma, Sumitomo, Sunovion, Taisho Pharmaceutical, Takeda, TC MSO, and Xhale; he has served on scientific advisory boards for the American Foundation for Suicide Prevention (AFSP), the Anxiety Disorders Association of America (ADAA), Bracket (Clintara), the Brain and Behavior Research Foundation, the Laureate Institute for Brain Research, Skyland Trail, and Xhale and on directorial boards for ADAA, AFSP, and Gratitude America; he is a stockholder in AbbVie, Antares, BI Gen Holdings, Celgene, Corcept Therapeutics, EMA Wellness, OPKO Health, Seattle Genetics, and Xhale; he receives income or has equity of $10,000 or more from American Psychiatric Publishing, Bracket (Clintara), CME Outfitters, Intra-Cellular Therapies, Magstim, Signant Health, Takeda, and Xhale; and he holds patents on a method and devices for transdermal delivery of lithium (patent 6,375,990B1) and a method of assessing antidepressant drug therapy via transport inhibition of monoamine neurotransmitters by ex vivo assay (patent 7,148,027B2).

References

1.
FDA-NIH Biomarker Working Group : BEST (Biomarkers, Endpoints, and Other Tools) Resource. Silver Spring and Bethesda, Md, US Food and Drug Administration and National Institutes of Health, 2016 ( https://www.ncbi.nlm.nih.gov/books/n/biomarkers/pdf/ )
2.
Abi-Dargham A, Horga G : The search for imaging biomarkers in psychiatric disorders. Nat Med 2016 ; 22 : 1248 – 1255
3.
Márquez F, Yassa MA : Neuroimaging biomarkers for Alzheimer’s disease. Mol Neurodegener 2019 ; 14 : 21
4.
Kraemer HC, Schultz SK, Arndt S : Biomarkers in psychiatry: methodological issues. Am J Geriatr Psychiatry 2002 ; 10 : 653 – 659
5.
First M, Botteron K, Carter C, et al: Consensus Report of the APA Work Group on Neuroimaging Markers of Psychiatric Disorders. Washington, DC, American Psychiatric Association, 2012
6.
Bough KJ, Amur S, Lao G, et al : Biomarkers for the development of new medications for cocaine dependence. Neuropsychopharmacology 2014 ; 39 : 202 – 219
7.
Park JW, Kerbel RS, Kelloff GJ, et al : Rationale for biomarkers and surrogate end points in mechanism-driven oncology drug development. Clin Cancer Res 2004 ; 10 : 3885 – 3896
8.
Tregellas JR : Neuroimaging biomarkers for early drug development in schizophrenia. Biol Psychiatry 2014 ; 76 : 111 – 119
9.
Breier A, Su TP, Saunders R, et al : Schizophrenia is associated with elevated amphetamine-induced synaptic dopamine concentrations: evidence from a novel positron emission tomography method. Proc Natl Acad Sci USA 1997 ; 94 : 2569 – 2574
10.
Hietala J, Syvälahti E : Dopamine in schizophrenia. Ann Med 1996 ; 28 : 557 – 561
11.
Seeman P, Kapur S : Schizophrenia: more dopamine, more D2 receptors. Proc Natl Acad Sci USA 2000 ; 97 : 7673 – 7675
12.
Lieberman JA, Kane JM, Alvir J : Provocative tests with psychostimulant drugs in schizophrenia. Psychopharmacology (Berl) 1987 ; 91 : 415 – 433
13.
Jones CA, Watson DJ, Fone KC : Animal models of schizophrenia. Br J Pharmacol 2011 ; 164 : 1162 – 1194
14.
Laruelle M, Abi-Dargham A, Gil R, et al : Increased dopamine transmission in schizophrenia: relationship to illness phases. Biol Psychiatry 1999 ; 46 : 56 – 72
15.
Kapur S, Zipursky R, Jones C, et al : Relationship between dopamine D(2) occupancy, clinical response, and side effects: a double-blind PET study of first-episode schizophrenia. Am J Psychiatry 2000 ; 157 : 514 – 520
16.
Nordström AL, Farde L, Wiesel FA, et al : Central D2-dopamine receptor occupancy in relation to antipsychotic drug effects: a double-blind PET study of schizophrenic patients. Biol Psychiatry 1993 ; 33 : 227 – 235
17.
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 – 8109
18.
Olney JW, Farber NB : Glutamate receptor dysfunction and schizophrenia. Arch Gen Psychiatry 1995 ; 52 : 998 – 1007
19.
Javitt DC, Zukin SR : Recent advances in the phencyclidine model of schizophrenia. Am J Psychiatry 1991 ; 148 : 1301 – 1308
20.
Moghaddam B, Javitt D : From revolution to evolution: the glutamate hypothesis of schizophrenia and its implication for treatment. Neuropsychopharmacology 2012 ; 37 : 4 – 15
21.
Coyle JT : NMDA receptor and schizophrenia: a brief history. Schizophr Bull 2012 ; 38 : 920 – 926
22.
Moghaddam B, Adams B, Verma A, et al : Activation of glutamatergic neurotransmission by ketamine: a novel step in the pathway from NMDA receptor blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex. J Neurosci 1997 ; 17 : 2921 – 2927
23.
Kegeles LS, Mao X, Stanford AD, et al : Elevated prefrontal cortex γ-aminobutyric acid and glutamate-glutamine levels in schizophrenia measured in vivo with proton magnetic resonance spectroscopy. Arch Gen Psychiatry 2012 ; 69 : 449 – 459
24.
de la Fuente-Sandoval C, León-Ortiz P, Azcárraga M, et al : Glutamate levels in the associative striatum before and after 4 weeks of antipsychotic treatment in first-episode psychosis: a longitudinal proton magnetic resonance spectroscopy study. JAMA Psychiatry 2013 ; 70 : 1057 – 1066
25.
Kraguljac NV, Morgan CJ, Reid MA, et al : A longitudinal magnetic resonance spectroscopy study investigating effects of risperidone in the anterior cingulate cortex and hippocampus in schizophrenia. Schizophr Res 2019 ; 210 : 239 – 244
26.
Kraguljac NV, White DM, Reid MA, et al : Increased hippocampal glutamate and volumetric deficits in unmedicated patients with schizophrenia. JAMA Psychiatry 2013 ; 70 : 1294 – 1302
27.
Plitman E, de la Fuente-Sandoval C, Reyes-Madrigal F, et al : Elevated myo-inositol, choline, and glutamate levels in the associative striatum of antipsychotic-naive patients with first-episode psychosis: a proton magnetic resonance spectroscopy study with implications for glial dysfunction. Schizophr Bull 2016 ; 42 : 415 – 424
28.
Marsman A, van den Heuvel MP, Klomp DW, et al : Glutamate in schizophrenia: a focused review and meta-analysis of 1H-MRS studies. Schizophr Bull 2013 ; 39 : 120 – 129
29.
Merritt K, Egerton A, Kempton MJ, et al : Nature of glutamate alterations in schizophrenia: a meta-analysis of proton magnetic resonance spectroscopy studies. JAMA Psychiatry 2016 ; 73 : 665 – 674
30.
Jackson ME, Homayoun H, Moghaddam B : NMDA receptor hypofunction produces concomitant firing rate potentiation and burst activity reduction in the prefrontal cortex. Proc Natl Acad Sci USA 2004 ; 101 : 8467 – 8472
31.
Lahti AC, Weiler MA, Tamara Michaelidis BA, et al : Effects of ketamine in normal and schizophrenic volunteers. Neuropsychopharmacology 2001 ; 25 : 455 – 467
32.
Kraguljac NV, Carle M, Frölich MA, et al : Mnemonic discrimination deficits in first-episode psychosis and a ketamine model suggests dentate gyrus pathology linked to N-methyl-d-aspartate receptor hypofunction. Biol Psychiatry Cogn Neurosci Neuroimaging 2018 ; 3 : 231 – 238
33.
Krystal JH, Karper LP, Seibyl JP, et al : Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans: psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry 1994 ; 51 : 199 – 214
34.
Rowland LM, Bustillo JR, Mullins PG, et al : Effects of ketamine on anterior cingulate glutamate metabolism in healthy humans: a 4-T proton MRS study. Am J Psychiatry 2005 ; 162 : 394 – 396
35.
Kraguljac NV, Frölich MA, Tran S, et al : Ketamine modulates hippocampal neurochemistry and functional connectivity: a combined magnetic resonance spectroscopy and resting-state fMRI study in healthy volunteers. Mol Psychiatry 2017 ; 22 : 562 – 569
36.
Stone JM, Dietrich C, Edden R, et al : Ketamine effects on brain GABA and glutamate levels with 1H-MRS: relationship to ketamine-induced psychopathology. Mol Psychiatry 2012 ; 17 : 664 – 665
37.
van der Aart J, Golla SSV, van der Pluijm M, et al : First in human evaluation of [18F]PK-209, a PET ligand for the ion channel binding site of NMDA receptors. EJNMMI Res 2018 ; 8 : 69
38.
Heckers S, Konradi C : GABAergic mechanisms of hippocampal hyperactivity in schizophrenia. Schizophr Res 2015 ; 167 : 4 – 11
39.
Lieberman JA, Girgis RR, Brucato G, et al : Hippocampal dysfunction in the pathophysiology of schizophrenia: a selective review and hypothesis for early detection and intervention. Mol Psychiatry 2018 ; 23 : 1764 – 1772
40.
Talati P, Rane S, Skinner J, et al : Increased hippocampal blood volume and normal blood flow in schizophrenia. Psychiatry Res 2015 ; 232 : 219 – 225
41.
Medoff DR, Holcomb HH, Lahti AC, et al : Probing the human hippocampus using rCBF: contrasts in schizophrenia. Hippocampus 2001 ; 11 : 543 – 550
42.
Tregellas JR, Smucny J, Harris JG, et al : Intrinsic hippocampal activity as a biomarker for cognition and symptoms in schizophrenia. Am J Psychiatry 2014 ; 171 : 549 – 556
43.
Molina V, Sanz J, Sarramea F, et al : Prefrontal atrophy in first episodes of schizophrenia associated with limbic metabolic hyperactivity. J Psychiatr Res 2005 ; 39 : 117 – 127
44.
Lahti AC, Weiler MA, Holcomb HH, et al : Correlations between rCBF and symptoms in two independent cohorts of drug-free patients with schizophrenia. Neuropsychopharmacology 2006 ; 31 : 221 – 230
45.
Lahti AC, Weiler MA, Holcomb HH, et al : Modulation of limbic circuitry predicts treatment response to antipsychotic medication: a functional imaging study in schizophrenia. Neuropsychopharmacology 2009 ; 34 : 2675 – 2690
46.
Schobel SA, Chaudhury NH, Khan UA, et al : Imaging patients with psychosis and a mouse model establishes a spreading pattern of hippocampal dysfunction and implicates glutamate as a driver. Neuron 2013 ; 78 : 81 – 93
47.
Lodge DJ, Grace AA : Hippocampal dysregulation of dopamine system function and the pathophysiology of schizophrenia. Trends Pharmacol Sci 2011 ; 32 : 507 – 513
48.
Dalmau J, Gleichman AJ, Hughes EG, et al : Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol 2008 ; 7 : 1091 – 1098
49.
Howes OD, McCutcheon R : Inflammation and the neural diathesis-stress hypothesis of schizophrenia: a reconceptualization. Transl Psychiatry 2017 ; 7 : e1024
50.
Mattei D, Schweibold R, Wolf SA : Brain in flames: animal models of psychosis: utility and limitations. Neuropsychiatr Dis Treat 2015 ; 11 : 1313 – 1329
51.
Trépanier MO, Hopperton KE, Mizrahi R, et al : Postmortem evidence of cerebral inflammation in schizophrenia: a systematic review. Mol Psychiatry 2016 ; 21 : 1009 – 1026
52.
Gallego JA, Blanco EA, Husain-Krautter S, et al : Cytokines in cerebrospinal fluid of patients with schizophrenia spectrum disorders: new data and an updated meta-analysis. Schizophr Res 2018 ; 202 : 64 – 71
53.
Marques TR, Ashok AH, Pillinger T, et al : Neuroinflammation in schizophrenia: meta-analysis of in vivo microglial imaging studies. Psychol Med 2019 ; 49 : 2186 – 2196
54.
Hafizi S, Tseng HH, Rao N, et al : Imaging microglial activation in untreated first-episode psychosis: a PET study with [18F]FEPPA. Am J Psychiatry 2017 ; 174 : 118 – 124
55.
van Berckel BN, Bossong MG, Boellaard R, et al : Microglia activation in recent-onset schizophrenia: a quantitative (R)-[11C]PK11195 positron emission tomography study. Biol Psychiatry 2008 ; 64 : 820 – 822
56.
Doorduin J, de Vries EF, Willemsen AT, et al : Neuroinflammation in schizophrenia-related psychosis: a PET study. J Nucl Med 2009 ; 50 : 1801 – 1807
57.
Takano A, Arakawa R, Ito H, et al : Peripheral benzodiazepine receptors in patients with chronic schizophrenia: a PET study with [11C]DAA1106. Int J Neuropsychopharmacol 2010 ; 13 : 943 – 950
58.
Kenk M, Selvanathan T, Rao N, et al : Imaging neuroinflammation in gray and white matter in schizophrenia: an in-vivo PET study with [18F]-FEPPA. Schizophr Bull 2015 ; 41 : 85 – 93
59.
Oestreich LK, Lyall AE, Pasternak O, et al : Characterizing white matter changes in chronic schizophrenia: a free-water imaging multi-site study. Schizophr Res 2017 ; 189:153–161
60.
Oestreich LK, Pasternak O, Shenton ME, et al : Abnormal white matter microstructure and increased extracellular free-water in the cingulum bundle associated with delusions in chronic schizophrenia. Neuroimage Clin 2016 ; 12 : 405 – 414
61.
Lyall AE, Pasternak O, Robinson DG, et al : Greater extracellular free-water in first-episode psychosis predicts better neurocognitive functioning. Mol Psychiatry 2018 ; 23:701–707
62.
Kraguljac NV, Anthony T, Monroe WS, et al : A longitudinal neurite and free water imaging study in patients with a schizophrenia spectrum disorder. Neuropsychopharmacology 2019 ; 44 : 1932 – 1939
63.
Pettersson-Yeo W, Allen P, Benetti S, et al : Dysconnectivity in schizophrenia: where are we now? Neurosci Biobehav Rev 2011 ; 35 : 1110 – 1124
64.
van den Heuvel MP, Fornito A : Brain networks in schizophrenia. Neuropsychol Rev 2014 ; 24 : 32 – 48
65.
Baker JT, Holmes AJ, Masters GA, et al : Disruption of cortical association networks in schizophrenia and psychotic bipolar disorder. JAMA Psychiatry 2014 ; 71 : 109 – 118
66.
Friston K, Brown HR, Siemerkus J, et al : The dysconnection hypothesis (2016). Schizophr Res 2016 ; 176 : 83 – 94
67.
Anticevic A, Cole MW, Repovs G, et al : Connectivity, pharmacology, and computation: toward a mechanistic understanding of neural system dysfunction in schizophrenia. Front Psychiatry 2013 ; 4 : 169
68.
Palaniyappan L, Simmonite M, White TP, et al : Neural primacy of the salience processing system in schizophrenia. Neuron 2013 ; 79 : 814 – 828
69.
Hadley JA, Kraguljac NV, White DM, et al : Change in brain network topology as a function of treatment response in schizophrenia: a longitudinal resting-state fMRI study using graph theory. NPJ Schizophr 2016 ; 2 : 16014
70.
Kraguljac NV, White DM, Hadley JA, et al : Abnormalities in large scale functional networks in unmedicated patients with schizophrenia and effects of risperidone. Neuroimage Clin 2015 ; 10 : 146 – 158
71.
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
72.
Driesen NR, McCarthy G, Bhagwagar Z, et al : Relationship of resting brain hyperconnectivity and schizophrenia-like symptoms produced by the NMDA receptor antagonist ketamine in humans. Mol Psychiatry 2013 ; 18 : 1199 – 1204
73.
Fleming LM, Javitt DC, Carter CS, et al : A multicenter study of ketamine effects on functional connectivity: large scale network relationships, hubs, and symptom mechanisms. Neuroimage Clin 2019 ; 22 : 101739
74.
Carletti F, Woolley JB, Bhattacharyya S, et al : Alterations in white matter evident before the onset of psychosis. Schizophr Bull 2012 ; 38 : 1170 – 1179
75.
Bloemen OJ, de Koning MB, Schmitz N, et al : White-matter markers for psychosis in a prospective ultra-high-risk cohort. Psychol Med 2010 ; 40 : 1297 – 1304
76.
Karlsgodt KH, Niendam TA, Bearden CE, et al : White matter integrity and prediction of social and role functioning in subjects at ultra-high risk for psychosis. Biol Psychiatry 2009 ; 66 : 562 – 569
77.
Kochunov P, Hong LE : Neurodevelopmental and neurodegenerative models of schizophrenia: white matter at the center stage. Schizophr Bull 2014 ; 40 : 721 – 728
78.
Xu H, Li XM : White matter abnormalities and animal models examining a putative role of altered white matter in schizophrenia. Schizophr Res Treatment 2011 ; 2011 : 826976
79.
Zhan Y, Paolicelli RC, Sforazzini F, et al : Deficient neuron-microglia signaling results in impaired functional brain connectivity and social behavior. Nat Neurosci 2014 ; 17 : 400 – 406
80.
Wolkin A, Choi SJ, Szilagyi S, et al : Inferior frontal white matter anisotropy and negative symptoms of schizophrenia: a diffusion tensor imaging study. Am J Psychiatry 2003 ; 160 : 572 – 574
81.
Skelly LR, Calhoun V, Meda SA, et al : Diffusion tensor imaging in schizophrenia: relationship to symptoms. Schizophr Res 2008 ; 98 : 157 – 162
82.
Bopp MHA, Zöllner R, Jansen A, et al : White matter integrity and symptom dimensions of schizophrenia: a diffusion tensor imaging study. Schizophr Res 2017 ; 184 : 59 – 68
83.
Kochunov P, Rowland LM, Fieremans E, et al : Diffusion-weighted imaging uncovers likely sources of processing-speed deficits in schizophrenia. Proc Natl Acad Sci USA 2016 ; 113 : 13504 – 13509
84.
Nestor PG, Kubicki M, Niznikiewicz M, et al : Neuropsychological disturbance in schizophrenia: a diffusion tensor imaging study. Neuropsychology 2008 ; 22 : 246 – 254
85.
Reis Marques T, Taylor H, Chaddock C, et al : White matter integrity as a predictor of response to treatment in first episode psychosis. Brain 2014 ; 137 : 172 – 182
86.
Mitelman SA, Newmark RE, Torosjan Y, et al : White matter fractional anisotropy and outcome in schizophrenia. Schizophr Res 2006 ; 87 : 138 – 159
87.
Luck D, Buchy L, Czechowska Y, et al : Fronto-temporal disconnectivity and clinical short-term outcome in first episode psychosis: a DTI-tractography study. J Psychiatr Res 2011 ; 45 : 369 – 377
88.
Cropley VL, Klauser P, Lenroot RK, et al : Accelerated gray and white matter deterioration with age in schizophrenia. Am J Psychiatry 2017 ; 174 : 286 – 295
89.
Gur RE, Turetsky BI, Bilker WB, et al : Reduced gray matter volume in schizophrenia. Arch Gen Psychiatry 1999 ; 56 : 905 – 911
90.
Thompson PM, Vidal C, Giedd JN, et al : Mapping adolescent brain change reveals dynamic wave of accelerated gray matter loss in very early-onset schizophrenia. Proc Natl Acad Sci USA 2001 ; 98 : 11650 – 11655
91.
Doostdar N, Kim E, Grayson B, et al : Global brain volume reductions in a sub-chronic phencyclidine animal model for schizophrenia and their relationship to recognition memory. J Psychopharmacol 2019 ; 33 : 1274 – 1287
92.
Barnes SA, Sawiak SJ, Caprioli D, et al : Impaired limbic cortico-striatal structure and sustained visual attention in a rodent model of schizophrenia. Int J Neuropsychopharmacol 2014 ; 18 : pyu010
93.
Crum WR, Sawiak SJ, Chege W, et al : Evolution of structural abnormalities in the rat brain following in utero exposure to maternal immune activation: a longitudinal in vivo MRI study. Brain Behav Immun 2017 ; 63 : 50 – 59
94.
Dempster K, Norman R, Théberge J, et al : Cognitive performance is associated with gray matter decline in first-episode psychosis. Psychiatry Res Neuroimaging 2017 ; 264 : 46 – 51
95.
Wible CG, Anderson J, Shenton ME, et al : Prefrontal cortex, negative symptoms, and schizophrenia: an MRI study. Psychiatry Res 2001 ; 108 : 65 – 78
96.
Padmanabhan JL, Tandon N, Haller CS, et al : Correlations between brain structure and symptom dimensions of psychosis in schizophrenia, schizoaffective, and psychotic bipolar I disorders. Schizophr Bull 2015 ; 41 : 154 – 162
97.
van Haren NE, Hulshoff Pol HE, Schnack HG, et al : Progressive brain volume loss in schizophrenia over the course of the illness: evidence of maturational abnormalities in early adulthood. Biol Psychiatry 2008 ; 63 : 106 – 113
98.
Steullet P, Cabungcal JH, Coyle J, et al : Oxidative stress-driven parvalbumin interneuron impairment as a common mechanism in models of schizophrenia. Mol Psychiatry 2017 ; 22 : 936 – 943
99.
Gonzalez-Burgos G, Fish KN, Lewis DA : GABA neuron alterations, cortical circuit dysfunction, and cognitive deficits in schizophrenia. Neural Plast 2011 ; 2011 : 723184
100.
Fusar-Poli P, Bonoldi I, Yung AR, et al : Predicting psychosis: meta-analysis of transition outcomes in individuals at high clinical risk. Arch Gen Psychiatry 2012 ; 69 : 220 – 229
101.
Koutsouleris N, Riecher-Rössler A, Meisenzahl EM, et al : Detecting the psychosis prodrome across high-risk populations using neuroanatomical biomarkers. Schizophr Bull 2015 ; 41 : 471 – 482
102.
Schobel SA, Lewandowski NM, Corcoran CM, et al : Differential targeting of the CA1 subfield of the hippocampal formation by schizophrenia and related psychotic disorders. Arch Gen Psychiatry 2009 ; 66 : 938 – 946
103.
Egerton A, Stone JM, Chaddock CA, et al : Relationship between brain glutamate levels and clinical outcome in individuals at ultra high risk of psychosis. Neuropsychopharmacology 2014 ; 39 : 2891 – 2899
104.
de la Fuente-Sandoval C, León-Ortiz P, Azcárraga M, et al : Striatal glutamate and the conversion to psychosis: a prospective 1H-MRS imaging study. Int J Neuropsychopharmacol 2013 ; 16 : 471 – 475
105.
Bossong MG, Antoniades M, Azis M, et al : Association of hippocampal glutamate levels with adverse outcomes in individuals at clinical high risk for psychosis. JAMA Psychiatry 2019 ; 76:199–207
106.
Kambeitz J, Kambeitz-Ilankovic L, Leucht S, et al : Detecting neuroimaging biomarkers for schizophrenia: a meta-analysis of multivariate pattern recognition studies. Neuropsychopharmacology 2015 ; 40 : 1742 – 1751
107.
Ulloa A, Plis S, Calhoun V: Improving Classification Rate of Schizophrenia Using a Multimodal Multi-Layer Perceptron Model with Structural and Functional MR2018. https://arxiv.org/abs/1804.04591
108.
Kegeles LS, Abi-Dargham A, Frankle WG, et al : Increased synaptic dopamine function in associative regions of the striatum in schizophrenia. Arch Gen Psychiatry 2010 ; 67 : 231 – 239
109.
Frankle WG, Gil R, Hackett E, et al : Occupancy of dopamine D2 receptors by the atypical antipsychotic drugs risperidone and olanzapine: theoretical implications. Psychopharmacology (Berl) 2004 ; 175 : 473 – 480
110.
Javitt DC, Carter CS, Krystal JH, et al : Utility of imaging-based biomarkers for glutamate-targeted drug development in psychotic disorders: a randomized clinical trial. JAMA Psychiatry 2018 ; 75 : 11 – 19
111.
Weickert CS, Weickert TW, Pillai A, et al : Biomarkers in schizophrenia: a brief conceptual consideration. Dis Markers 2013 ; 35 : 3 – 9
112.
Bodnar M, Harvey PO, Malla AK, et al : The parahippocampal gyrus as a neural marker of early remission in first-episode psychosis: a voxel-based morphometry study. Clin Schizophr Relat Psychoses 2011 ; 4 : 217 – 228
113.
Li M, Li X, Das TK, et al : Prognostic utility of multivariate morphometry in schizophrenia. Front Psychiatry 2019 ; 10 : 245
114.
Palaniyappan L, Liddle PF : Aberrant cortical gyrification in schizophrenia: a surface-based morphometry study. J Psychiatry Neurosci 2012 ; 37 : 399 – 406
115.
Nelson EA, White DM, Kraguljac NV, et al : Gyrification connectomes in unmedicated patients with schizophrenia and following a short course of antipsychotic drug treatment. Front Psychiatry 2018 ; 9 : 699
116.
Palaniyappan L, Marques TR, Taylor H, et al : Cortical folding defects as markers of poor treatment response in first-episode psychosis. JAMA Psychiatry 2013 ; 70 : 1031 – 1040
117.
Egerton A, Broberg BV, Van Haren N, et al : Response to initial antipsychotic treatment in first episode psychosis is related to anterior cingulate glutamate levels: a multicentre 1H-MRS study (OPTiMiSE). Mol Psychiatry 2018 ; 23 : 2145 – 2155
118.
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 – 752
119.
Egerton A, Bhachu A, Merritt K, et al : Effects of antipsychotic administration on brain glutamate in schizophrenia: a systematic review of longitudinal (1)H-MRS studies. Front Psychiatry 2017 ; 8 : 66
120.
Sarpal DK, Robinson DG, Lencz T, et al : Antipsychotic treatment and functional connectivity of the striatum in first-episode schizophrenia. JAMA Psychiatry 2015 ; 72 : 5 – 13
121.
White TP, Wigton R, Joyce DW, et al : Dysfunctional striatal systems in treatment-resistant schizophrenia. Neuropsychopharmacology 2016 ; 41 : 1274 – 1285
122.
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 – 1055
123.
Hadley JA, Nenert R, Kraguljac NV, et al : Ventral tegmental area/midbrain functional connectivity and response to antipsychotic medication in schizophrenia. Neuropsychopharmacology 2014 ; 39 : 1020 – 1030
124.
Doucet GE, Moser DA, Luber MJ, et al : Baseline brain structural and functional predictors of clinical outcome in the early course of schizophrenia. Mol Psychiatry 2020 ; 25:863–872
125.
Wang LX, Guo F, Zhu YQ, et al : Effect of second-generation antipsychotics on brain network topology in first-episode schizophrenia: a longitudinal rs-fMRI study. Schizophr Res 2019 ; 208 : 160 – 166
126.
Cao B, Cho RY, Chen D, et al : Treatment response prediction and individualized identification of first-episode drug-naive schizophrenia using brain functional connectivity. Mol Psychiatry 2020 ; 25:906–913
127.
Cui LB, Cai M, Wang XR, et al : Prediction of early response to overall treatment for schizophrenia: a functional magnetic resonance imaging study. Brain Behav 2019 ; 9 : e01211
128.
Cuthbert BN, Insel TR : Toward the future of psychiatric diagnosis: the seven pillars of RDoC. BMC Med 2013 ; 11 : 126
129.
Insel TR : The NIMH Research Domain Criteria (RDoC) Project: precision medicine for psychiatry. Am J Psychiatry 2014 ; 171 : 395 – 397
130.
Tamminga CA, Pearlson GD, Stan AD, et al : Strategies for advancing disease definition using biomarkers and genetics: the bipolar and schizophrenia network for intermediate phenotypes. Biol Psychiatry Cogn Neurosci Neuroimaging 2017 ; 2 : 20 – 27
131.
Goff DC, Romero K, Paul J, et al : Biomarkers for drug development in early psychosis: current issues and promising directions. Eur Neuropsychopharmacol 2016 ; 26 : 923 – 937
132.
Glantz LA, Lewis DA : Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia. Arch Gen Psychiatry 2000 ; 57 : 65 – 73
133.
Berdenis van Berlekom A, Muflihah CH, Snijders GJLJ, et al : Synapse pathology in schizophrenia: a meta-analysis of postsynaptic elements in postmortem brain studies. Schizophr Bull 2020 ; 46 : 374 – 386
134.
Grzenda A, Widge AS : Electroencephalographic biomarkers for predicting antidepressant response: new methods, old questions. JAMA Psychiatry 2020 ; 77 : 347 – 348
135.
Widge AS, Bilge MT, Montana R, et al : Electroencephalographic biomarkers for treatment response prediction in major depressive illness: a meta-analysis. Am J Psychiatry 2019 ; 176 : 44 – 56
136.
Pletcher MJ, Pignone M : Evaluating the clinical utility of a biomarker: a review of methods for estimating health impact. Circulation 2011 ; 123 : 1116 – 1124
137.
Parkinson DR, McCormack RT, Keating SM, et al : Evidence of clinical utility: an unmet need in molecular diagnostics for patients with cancer. Clin Cancer Res 2014 ; 20 : 1428 – 1444
138.
Insel TR, Landis SC, Collins FS ; The NIH BRAIN Initiative : Research priorities. Science 2013 ; 340 : 687 – 688
139.
Gosho M, Nagashima K, Sato Y : Study designs and statistical analyses for biomarker research. Sensors (Basel) 2012 ; 12 : 8966 – 8986
140.
Makowski C, Lepage M, Evans AC : Head motion: the dirty little secret of neuroimaging in psychiatry. J Psychiatry Neurosci 2019 ; 44 : 62 – 68
141.
Cetin Karayumak S, Bouix S, Ning L, et al : Retrospective harmonization of multi-site diffusion MRI data acquired with different acquisition parameters. Neuroimage 2019 ; 184 : 180 – 200
142.
Fusar-Poli P, Hijazi Z, Stahl D, et al : The science of prognosis in psychiatry: a review. JAMA Psychiatry 2018 ; 75 : 1289 – 1297
143.
Calhoun VD, Lawrie SM, Mourao-Miranda J, et al : Prediction of individual differences from neuroimaging data. Neuroimage 2017 ; 145 ( Pt B ): 135 – 136
144.
Davatzikos C : Machine learning in neuroimaging: progress and challenges. Neuroimage 2019 ; 197 : 652 – 656
145.
Woo CW, Chang LJ, Lindquist MA, et al : Building better biomarkers: brain models in translational neuroimaging. Nat Neurosci 2017 ; 20 : 365 – 377
146.
National Institute of Mental Health: Explainable artificial intelligence for decoding and modulating neural circuit activity linked to behavior. 2019. https://grants.nih.gov/grants/guide/pa-files/PAR-19-344.html
147.
Pepe MS, Janes H, Li CI, et al : Early-phase studies of biomarkers: what target sensitivity and specificity values might confer clinical utility? Clin Chem 2016 ; 62 : 737 – 742
148.
Pepe MS, Feng Z, Janes H, et al : Pivotal evaluation of the accuracy of a biomarker used for classification or prediction: standards for study design. J Natl Cancer Inst 2008 ; 100 : 1432 – 1438
149.
Poline JB, Breeze JL, Ghosh S, et al : Data sharing in neuroimaging research. Front Neuroinform 2012 ; 6 : 9
150.
Schulz KF, Altman DG, Moher D ; CONSORT Group : CONSORT 2010 statement: updated guidelines for reporting parallel group randomised trials. BMJ 2010 ; 340 : c332
151.
Moher D, Liberati A, Tetzlaff J, et al : Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. BMJ 2009 ; 339 : b2535
152.
Scherer A : Reproducibility in biomarker research and clinical development: a global challenge. Biomarkers Med 2017 ; 11 : 309 – 312
153.
Weiner MW, Veitch DP, Aisen PS, et al : Impact of the Alzheimer’s Disease Neuroimaging Initiative, 2004 to 2014. Alzheimers Dement 2015 ; 11 : 865 – 884

Information & Authors

Information

Published In

Go to American Journal of Psychiatry
Go to American Journal of Psychiatry
American Journal of Psychiatry
Pages: 509 - 521
PubMed: 33397140

History

Received: 26 March 2020
Revision received: 5 July 2020
Accepted: 5 October 2020
Published online: 5 January 2021
Published in print: June 2021

Keywords

  1. Neuroimaging
  2. Biomarker
  3. Schizophrenia Spectrum and Other Psychotic Disorders
  4. Work Group on Biomarkers and Novel Treatments of the APA Council of Research

Authors

Details

Nina V. Kraguljac, M.D. [email protected]
Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham (Kraguljac); Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (McDonald); Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis (Widge); Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, Calif., and Veterans Affairs Palo Alto Health Care System, Palo Alto, Calif. (Rodriguez); Department of Psychiatry and Behavioral Sciences, University of New Mexico Health Sciences Center, Albuquerque (Tohen); Department of Psychiatry, University of Texas Dell Medical School, Austin (Nemeroff).
William M. McDonald, M.D.
Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham (Kraguljac); Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (McDonald); Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis (Widge); Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, Calif., and Veterans Affairs Palo Alto Health Care System, Palo Alto, Calif. (Rodriguez); Department of Psychiatry and Behavioral Sciences, University of New Mexico Health Sciences Center, Albuquerque (Tohen); Department of Psychiatry, University of Texas Dell Medical School, Austin (Nemeroff).
Alik S. Widge, M.D., Ph.D.
Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham (Kraguljac); Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (McDonald); Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis (Widge); Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, Calif., and Veterans Affairs Palo Alto Health Care System, Palo Alto, Calif. (Rodriguez); Department of Psychiatry and Behavioral Sciences, University of New Mexico Health Sciences Center, Albuquerque (Tohen); Department of Psychiatry, University of Texas Dell Medical School, Austin (Nemeroff).
Carolyn I. Rodriguez, M.D., Ph.D.
Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham (Kraguljac); Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (McDonald); Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis (Widge); Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, Calif., and Veterans Affairs Palo Alto Health Care System, Palo Alto, Calif. (Rodriguez); Department of Psychiatry and Behavioral Sciences, University of New Mexico Health Sciences Center, Albuquerque (Tohen); Department of Psychiatry, University of Texas Dell Medical School, Austin (Nemeroff).
Mauricio Tohen, M.D., Dr.P.H.
Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham (Kraguljac); Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (McDonald); Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis (Widge); Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, Calif., and Veterans Affairs Palo Alto Health Care System, Palo Alto, Calif. (Rodriguez); Department of Psychiatry and Behavioral Sciences, University of New Mexico Health Sciences Center, Albuquerque (Tohen); Department of Psychiatry, University of Texas Dell Medical School, Austin (Nemeroff).
Charles B. Nemeroff, M.D., Ph.D.
Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham (Kraguljac); Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (McDonald); Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis (Widge); Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, Calif., and Veterans Affairs Palo Alto Health Care System, Palo Alto, Calif. (Rodriguez); Department of Psychiatry and Behavioral Sciences, University of New Mexico Health Sciences Center, Albuquerque (Tohen); Department of Psychiatry, University of Texas Dell Medical School, Austin (Nemeroff).

Notes

Send correspondence to Dr. Kraguljac ([email protected]).

Funding Information

Supported by NIMH grants K23MH106683 and R01MH118484 to Dr. Kraguljac.

Metrics & Citations

Metrics

Citations

Export Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click Download.

For more information or tips please see 'Downloading to a citation manager' in the Help menu.

Format
Citation style
Style
Copy to clipboard

View Options

View options

PDF/EPUB

View PDF/EPUB

Login options

Already a subscriber? Access your subscription through your login credentials or your institution for full access to this article.

Personal login Institutional Login Open Athens login
Purchase Options

Purchase this article to access the full text.

PPV Articles - American Journal of Psychiatry

PPV Articles - American Journal of Psychiatry

Not a subscriber?

Subscribe Now / Learn More

PsychiatryOnline subscription options offer access to the DSM-5-TR® library, books, journals, CME, and patient resources. This all-in-one virtual library provides psychiatrists and mental health professionals with key resources for diagnosis, treatment, research, and professional development.

Need more help? PsychiatryOnline Customer Service may be reached by emailing [email protected] or by calling 800-368-5777 (in the U.S.) or 703-907-7322 (outside the U.S.).

Media

Figures

Other

Tables

Share

Share

Share article link

Share