The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
ReviewsFull Access

A Clinician’s Guide to PTSD Biomarkers and Their Potential Future Use

Abstract

No clinically validated biomarkers have yet been found to assist in the diagnosis and treatment of posttraumatic stress disorder (PTSD). Innovation in clinical trial design, however, has led to the study of biomarkers as part of testing new medications and psychotherapies. There may soon be viable biomarkers to assist in diagnosis of PTSD and prediction of illness trajectory, severity, and functional outcomes; subtyping; and treatment selection. Processes for the identification and validation of biomarker findings are complex, involving several stages of clinical testing before use. The authors provide an overview of issues regarding the clinical use of PTSD biomarkers and examine a set of genetic, epigenetic, and other blood-based markers along with physiological markers currently proposed as candidate tests for PTSD. Studies that have identified candidate biomarkers with relevance to treatment selection in PTSD are discussed as a promising area of research that may lead to changes in clinical practice.

Posttraumatic stress disorder (PTSD), as defined in the DSM-5 (1), develops after exposure to a traumatic event. The DSM-5 identifies four PTSD symptom clusters: intrusion symptoms, persistent avoidance, negative alterations in cognitions and mood, and marked alterations in arousal and reactivity. Reflecting the challenges of clinical care and research, the list of potential symptoms comprises more than 600,000 ways to meet criteria for PTSD (2). The notable heterogeneity of the PTSD diagnosis suggests that biologically homogeneous groups—“biotypes” (3)—might exist as PTSD subtypes and might respond to different treatments, as we are now finding with depression (4).

The prospect of meaningful biomarkers for the assessment and treatment of PTSD has intuitive appeal as an enhancement of current clinical practice. There remain several questions, however. Which biomarkers are the current focus of research? How will they be used? When will they become available in routine clinical practice? PTSD is presently a clinical diagnosis, assessed through an interview and patient self-report. No validated biological measures or laboratory tests currently exist to confirm the diagnosis of PTSD, to monitor disease progression, to assess treatment response, or to select appropriate treatments for particular individuals.

In the context of advances in basic and clinical neuroscience over the past two decades, the identification of reliable biomarkers for PTSD has become an active area of research that is more promising than ever. Furthermore, during the wars in Iraq and Afghanistan, a pressing need for new ways to identify and treat psychological health conditions among military service members led to intensification of efforts to develop biological markers for PTSD (5, 6). The search for clinically relevant biomarkers is necessarily broad and encompasses blood-based assays as well as physiological and clinical measurements.

As discussed in greater detail below, a biomarker is generally defined as a measurable indicator of a biological process. A foundational understanding of the biological basis of PTSD is thus key to this line of research. Although a number of studies had previously described biological features of PTSD, a key discussion in 2004 identified a need for postmortem samples to directly examine in brain tissue the biological correlates of the diagnosis and for subtyping (7). This recognition of the importance of postmortem samples resulted in the successful establishment of several brain banks, in particular the National PTSD Brain Bank (8, 9), and large collections of blood and saliva samples for further study (5, 10, 11). Postmortem brain tissue will likely play a significant role in the identification of mechanisms and markers as well as subsequent characterization of peripheral markers and assay development.

With an emphasis on outlining the anticipated future state of biomarkers for clinical practice, we describe the different types of PTSD biomarkers that are potentially useful and some of the obstacles that the field may encounter as biomarkers are further developed for clinical use (see Box 1). We then discuss examples of promising blood-based and physiological markers of PTSD and point to a particularly relevant and evolving area of research, the use of biomarkers as predictors of treatment response.

BOX 1. KEY POINTS

• At this time, there are no clinically validated biomarkers to assist in the diagnosis and treatment of individuals with posttraumatic stress disorder (PTSD).

• A defined temporal relationship between trauma exposure and development of PTSD affords an opportunity to examine disease onset and clarify Gene × Environment interactions in PTSD and trauma-related disorders.

• There may soon be viable biomarkers to assist in the diagnosis of PTSD and the prediction of illness trajectory, severity, and functional outcomes, as well as for subtyping and treatment selection.

• Processes for the identification and validation of biomarker findings are complex, involving several stages of clinical testing prior to use.

• Understanding and anticipating the potential uses of PTSD biomarkers is essential for advancing the development of clinically viable tests.

Distinctive Features of PTSD

Precision medicine aims to identify individual-level phenomenological characteristics (phenotype), underlying biological correlates (endophenotype), and biological targets implicated in modifying the course of disease. The conceptual framework for biomarker development in PTSD begins with understanding the onset, progression, trajectories of illness, and process of recovery from PTSD. Biomarkers must be specific and linked to clinical phenotypes. Among DSM-5 disorders, PTSD is distinct as a psychological condition defined by exposure to a traumatic event at a particular point (or points) in time. As one of the few psychiatric diagnoses linked to a causal event, PTSD provides unique opportunities to study the time course of biological changes involved in the onset of psychiatric disease. As an exposure-related disorder, therefore, PTSD is a classic example of a Gene × Environment (G×E) interaction that may inform mechanisms of onset in other disorders.

The basis of psychiatric biomarkers is similar to biomarkers for nonpsychiatric disorders in that there are static markers (e.g., genetic testing for Huntington’s disease or BRCA1/BRCA2 for breast cancer) and ones that are quantitative and may change over time (e.g., HgA1c, thyroid-stimulating hormone). In PTSD, biomarkers can be examined over a time course: before and after trauma exposure and during the initial weeks to months after exposure. Similar to previously proposed typologies (12, 13), PTSD biomarkers may therefore include markers as indicated in Box 2.

BOX 2. TYPOLOGY OF PTSD MARKERS

• Risk: identification of who is at risk for posttraumatic stress disorder (PTSD) after trauma exposure–and when the individual is at greatest risk. There may also be biomarkers of risk for trauma exposure itself, which it is important to separate from the disease biomarkers.

• Vulnerability to specific types and intensities of trauma exposure: In PTSD, trauma types and intensity of exposure may differ widely. Individuals certainly differ in their vulnerability as well as resilience to various traumatic events. Similarly, people’s capacity for resilience in the face of trauma may be overwhelmed at different levels of exposure. For instance, multiple severe trauma exposures may override the capacity of a highly resilient individual, whereas a single, lower magnitude trauma may become significant for others.

• Onset of PTSD: detection of transition to psychopathology. The onset of disease may involve specific “onset” biological events that, once set into motion, are no longer part of the ongoing biological pathology. Therefore, such biomarkers may or may not have a relationship to the mechanisms of PTSD symptomatology or to treatment targets.

• PTSD diagnosis: identification of patients who meet full diagnostic criteria, and assessment of the severity of symptoms.

• Subtype diagnosis (“biotypes”): establishing biologically homogeneous phenotypes. One of the most promising sets of investigations involves subtyping according to symptom profile (e.g., relative predominance of certain symptoms) using biomarkers to further characterize biological characteristics related to certain symptom clusters. Alternatively, subtyping may be the result of noting specific clusters of neurobiological response, such as on functional magnetic resonance imaging.

• Prognosis: prediction of trajectory of illness and functional outcomes. Three types of prognostic information may be clinically useful: predictors of the level of symptom severity, predictors of the course of PTSD over time (recovery, late onset, chronic, resilience), and long-term outcomes. For instance, a set of biomarkers might identify a population of individuals who are likely to experience early severe symptoms with resolution over the first months of the illness, as distinguished from those with late onset and symptoms that do not resolve, resulting in persistent impairment.

• Phase of disease: identification of the individual’s current state relative to the expected course of illness. Markers may differ according to the phase of evolution or time since the trauma. Predictors of onset, early disease, chronic disease, and postdisease recovery may well be present. Baseline predictors (e.g., ones that are static—e.g., childhood adversity) may be distinct from acute markers of stress in the initial weeks after trauma. Diagnostic markers of psychopathology later in the time course (e.g., six to twelve months after exposure) may index long-term neurobiological changes that are undetectable in the setting of acute stress.

• Recovery and monitoring for treatment response: detection of resolution of the illness. Resolution may also have several different paths. Biomarkers may serve as an indicator that covaries with response to treatment if the marker is in the mechanistic pathway (a treatment target) or if it is a product of the disease process without serving as a locus for intervention.

• Treatment selection: optimization of treatment selection by matching of treatment to individual characteristics. Pretreatment markers may predict treatment response and thereby enhance the accuracy of treatment selection. This line of biomarker inquiry in particular may be the most effective for changing clinician behavior and improving patient outcomes.

Given the number of different types of biomarkers that can be investigated, an understanding of how the markers will influence clinical practice is essential to guide development. However, research evidence is not established according to the above types of biomarkers. We therefore summarize the current state of biomarker research with a focus on clinically relevant trends, keeping in mind the prospect of further differentiation of biomarkers according to their projected clinical use.

Why are there No Available or Soon-to-Be Available Biomarker Assays for PTSD?

The use of existing, commercially available measurements, such as a standard laboratory test (e.g., C-reactive protein, electroencephalography [EEG], or functional magnetic resonance imaging) may become feasible if research evidence supports their reliability and validity as markers for PTSD. In contrast, the development of novel chemical, proteomic, or genetic markers involves a longer process, given the additional validation needed for the introduction of a new test, prior to commercialization and clinical implementation.

As is true for other psychiatric disorders, diagnostic biomarkers for PTSD pose a particular difficulty in that there is no biological gold standard. Unlike a tumor, for which discrete tissue samples are available for comparison, PTSD is not readily localized in one brain region and may involve subtle alterations to physiologically normal brain circuits or connectivity. Animal models of PTSD, none of which model the entire disorder, are arguably homogenous by design, to represent and detect basic mechanisms. They have been used to screen for putative biomarkers, but this method of discovery has limitations, given minimal homology with human biology and the heterogeneity present in clinical populations.

The study of postmortem human brains may allow for more robust signal detection, followed by the development of methods to detect corresponding biomarkers in peripheral blood. Appropriate selection of clinical rating instruments and the analysis of dimensional clinical constructs in conjunction with biological findings may further assist in parsing the heterogeneity of PTSD. Combinations of clinical instruments with currently available and new assays in large datasets may be needed to fully characterize the heterogeneity among individuals with a PTSD diagnosis, and machine-learning methods may improve signal detection (14, 15).

Once screening and discovery have identified candidate biomarkers, analytic validation to enhance assay precision and accuracy, as well as to establish thresholds for detection, will be needed (16). Acceptable thresholds for precision, accuracy, and detection should be determined in accordance with the intended clinical use (16). As noted more generally for screening tests, the sensitivity, specificity, positive predictive value, and negative predictive value across a large, diverse population are highly consequential. With an accuracy level highly dependent on the base rate of the disease, laboratory tests can result in costly follow-on evaluations, which themselves may entail risks, additional costs, and inconvenience to the patient. Defining the context of use, including the target population, is therefore important to the biomarker development process, as well as the ways that a biomarker test may enhance the clinician’s practice (17).

Uses of PTSD Biomarkers

According to a recent glossary from the U.S. Food and Drug Administration and the National Institutes of Health, a biomarker is a “defined characteristic that is measured as an indicator of normal biological processes, pathogenic processes, or responses to an exposure or intervention, including therapeutic interventions. Molecular, histologic, radiographic, or physiological characteristics are types of biomarkers. A biomarker is not an assessment of how an individual feels, functions, or survives” (13). The term indicator suggests that a biomarker does not necessarily index a mechanistic pathway or serve as a target for intervention. A biomarker may be a characteristic that is correlated with disease or produced as the biological result of a disease process and thereby serve as a marker without a demonstrated causal relationship (18).

One notable example of the complexity in the clinical implementation of biomarkers is the medical community’s experience with prostate-specific antigen (PSA). Beginning in the 1980s, PSA was used as a standard screening measure for prostate cancer among older men. In the early 2000s, several large studies to determine the efficacy of this screening process (19) showed that universal screening resulted in substantial adverse outcomes from follow-up biopsies, whereas the number of prostate cancer deaths prevented was less than 1 in 1,000 men screened (20). Although PSA is no longer recommended for general screening among older men, it remains a useful tool in situations such as monitoring tumor recurrence for individuals previously treated for prostate cancer. This example from general medicine illustrates the point that when one is thinking through the clinical use of biomarkers, it is important to carefully consider the context of use and secondary effects of broad-based testing in contrast to tailored use within specified populations or known disease processes.

PTSD biomarkers thus far have been used exclusively in the research setting. As described above, PTSD has a specific event as its starting point, with the opportunity to examine evolution over time and distinguish trajectories of recovery, a process that is optimally studied with longitudinal data with repeated measures. A promising example of longitudinal design is a currently ongoing study funded by the National Institute of Mental Health involving 5,000 trauma-exposed participants recruited in an emergency room setting. This study will use biological markers for phenotyping and conduct surveys and continuous real-time physiological monitoring over eight weeks, with a 52-week total follow-up period (21). In addition, the research use of biomarkers over the course of interventional studies is well recognized, although clinical trials have generally recruited on the basis of a PTSD diagnosis, using biomarkers to define moderators and subgroups of responders. The collection of blood and saliva samples to be stored for future biomarker studies has become a common practice in clinical trials.

These research uses of biomarkers for PTSD set the stage for their future clinical use, although their implementation in clinical practice raises many difficult questions. The potential development of a test that will assist in establishing a PTSD categorical diagnosis has been considered. However, the clinician immediately faces an issue regarding the manner in which an assay can be used to enhance clinical judgment. Does a negative result necessarily lead to the conclusion that the individual does not have PTSD? A biomarker test might assist in screening for possible PTSD in a primary care setting for subsequent referral to specialty consultation. Alternatively, a biomarker assay in the specialty setting might be used to help establish or rule out the diagnosis in clinical presentations that are ambiguous, where the clinician might have concerns if the patient has a low level of awareness of symptoms and their affect on functioning.

The cost-benefit issues for different types of PTSD biomarkers have been discussed in detail elsewhere (12). Guidelines are needed to establish standards so that biomarkers are not used to supplant the clinician’s judgment or decrease the care with which clinical assessment is performed. In the future, there may well be biomarker-positive PTSD cases as well as biomarker-negative cases among individuals who nonetheless require treatment. The potential use of biological markers in legal and administrative contexts has yet to be discussed in detail and resolved.

For instance, the ethical use of biomarkers for decisions regarding recruitment of military service members, fitness for duty determinations for police officers, or examination of malingering in forensic settings requires a clear understanding and consensus regarding the benefits and limitations of particular tests. The level of accuracy needed to rely on biomarkers for decision making in these contexts would likely be very high. Using biomarkers to deny disability benefits or to disqualify individuals from military service would be ethically problematic from the perspectives of autonomy and potential harm as well as fairness to the overall population concerned.

Blood-Based Biomarkers

Biological markers identified in earlier investigations include heart rate, electrodermal response (skin conductance), cortisol levels, and glucocorticoid receptor (GR) density in peripheral blood cells (22). In the past decade, biomarker research has focused further on these, along with genetic and epigenetic markers, mitochondrial genes, mRNA, and micro RNA. A large number of chemical, protein, and genetic markers have been examined among individuals with PTSD (see (2224 and Box 2).

Glucocorticoid regulation in PTSD has been well characterized, and many potential markers pertain to glucocorticoid pathways (25). The hypothalamic-pituitary-adrenal (HPA) axis is a complex system that controls stress response. Although basal serum cortisol levels have demonstrated mixed results as a predictor or marker of PTSD, decreased cortisol reactivity to an experimental stressor was found to predict the development of PTSD (22). PTSD symptom severity has been associated with high pre-exposure GR number in peripheral blood mononuclear cells (2527) and T-cell sensitivity to glucocorticoids for PTSD participants without depression (28). Data also show that individuals with PTSD had elevation of cortisol (29) and corticotropin-releasing hormone (CRH) concentrations (30) in cerebrospinal fluid when compared with healthy controls. In contrast, CRH concentrations in cerebrospinal fluid seem to decrease after exposure to a trauma-related stimulus among individuals with PTSD (31). Other potential biomarkers include genetic variants of the CRH receptor (32) and lower methylation of a promotor region for the GR gene (33).

A number of biomarkers related to additional pathways involved in regulating the HPA axis have been proposed. FK506 binding protein 51 (FKBP5) is a heat-shock protein that binds to and regulates GR activity (34), with variants that are implicated in a number of psychiatric disorders, including PTSD (35). Among the numerous proteins that bind to the immunosuppressant drug FK506, functional polymorphisms in the gene-encoding FKBP5 have been associated with risk among adults of developing stress-related psychiatric disorders (34, 36), as well as with PTSD symptom severity (37). Carriers of one risk allele had GR supersensitivity and PTSD, which suggests that functional variants in FKBP5 are associated with biologically distinct subtypes of PTSD (38). The FKBP5 promoter region contains a glucocorticoid response element whose demethylation is associated with childhood exposure to trauma and may be linked to stress-dependent changes in gene transcription (39). The demethylation of the FKBP5 gene is associated with long-term dysregulation of the stress response and global immune cell functioning (39). Low FKBP5 mRNA among predeployment soldiers predicted more intense PTSD symptoms (27).

Also related to the HPA axis stress response in PTSD is the pituitary adenylate cyclase-activating polypeptide (PACAP). PACAP and its receptors have been identified in both autonomic stress and HPA pathways (40). Higher blood levels of PACAP have been associated with a PTSD diagnosis, symptom severity, and potentiated startle among female but not male participants (41). A further genetic study revealed that a polymorphism in the PACAP receptor gene, found within an estrogen response element, was associated with PTSD diagnosis among women and that differential methylation of this gene correlated with PTSD symptoms (41). Further supporting the role of stress-response dysregulation, expression of serum and glucocorticoid regulated kinase 1 was down-regulated in the prefrontal cortex (PFC) of postmortem samples from PTSD participants (42).

p11, a calcium-binding protein associated with PTSD as well as depression and regulated by glucocorticoids, has been found to be overexpressed in the postmortem brain of individuals with PTSD (43). In contrast, it is decreased in other psychiatric disorders, such as depression (44), which suggests that p11 may serve as specific biomarker for PTSD. The overexpression in PTSD is thought to be regulated by glucocorticoids associated with stress, and it results in a significant increase of GR activation. Activated GR is then transferred into the nucleus, where it interacts with glucocorticoid response elements in p11. Concomitantly, mRNA expression is decreased in PTSD, whereas it is elevated in major depressive disorder, bipolar disorder, and schizophrenia, which suggests that p11 may serve as a biomarker specific to trauma-related disorders (45).

Brain-derived neurotrophic factor (BDNF) is implicated in dendritic spine density, synaptic plasticity, and memory formation (46, 47) as well as in a number of psychiatric disorders (46, 48). Chronic exposure to stress is associated with elevated BDNF in the basolateral amygdala and decreased levels in the hippocampus (47). BDNF is modulated by mineralocorticoids and glucocorticoids, both implicated in traumatic stress reactions (47).

In a study of traffic accident survivors evaluated at six months, BDNF levels in plasma and the Val66Met genotype were not associated with PTSD, but plasma BDNF was correlated with more lifetime trauma exposures (49). In another study, PTSD diagnosis was associated with significantly lower plasma BDNF (50), although BDNF may be elevated early in the course of illness, decreasing with time (51). A single nucleotide polymorphism, Val66Met, has been implicated in susceptibility to a number of neuropsychiatric disorders, including PTSD (46). The frequency of at least one Met allele was twofold higher among individuals with probable PTSD than among controls, and plasma BDNF was also significantly higher among those with probable PTSD (52). Furthermore, the frequency of the BDNF Val66met polymorphism was significantly higher among participants with PTSD who also had an exaggerated startle reflex. In contrast, the frequency of the Val/Val genotype was higher among participants without PTSD and those with a low startle response, compared with those with PTSD and with a high startle response.

Data also suggest a G×E interaction such that at a lower level of exposure to lifetime stressors, the Met/Met genotype was associated with an elevated risk of hyperarousal, whereas Val allele carriers had a lower rate of hyperarousal. Among individuals who reported fewer lifetime stressors, the Met/Met genotype was more highly positively associated with the PTSD Checklist total score than were Val carriers. This suggests that at a lower level of stressful events, Met/Met genotype individuals had higher levels of PTSD symptoms, and Val carriers had lower levels of PTSD symptoms (52). This result contrasts with other studies of genetic association that showed a lack of relationship between the BDNF Val66Met allele and a PTSD diagnosis (49, 53, 54), possibly as a result of methodological differences. Although with further research BDNF may hold promise as a biomarker for trauma- and stressor-related disorders, BDNF is also altered in phobia and panic disorder (46) and other central nervous system disorders, including Huntington’s disease, Alzheimer’s disease, schizophrenia, depression, and substance abuse (48). BDNF involvement in a wide array of disorders suggests that its role in PTSD potentially involves common pathways or mechanisms.

Epigenetic regulation of genes through DNA methylation is a promising area of research for detection of PTSD-related alterations (55). Studies have predominantly used models in animals and human peripheral blood samples. Consistent with the impetus for a PTSD brain bank, Zannas et al. (56) reinforced that, given the neuroanatomical specificity of epigenetic modifications, human postmortem studies in brain tissue are crucial for improved detection.

In addition to the above, a number of other candidate biomarkers have been explored, encompassing a wide range of neuroendocrine, genetic, and inflammatory markers that has been well summarized elsewhere (22). These include 5-HTTLPR polymorphisms in the promoter region of the serotonin transporter gene (SLC6A4) (57). These studies suggest that among European Americans there is an interactive effect of childhood exposure and SLC6A4 gene alleles on risk for PTSD.

In studies of serum in participants with PTSD and controls among U.S. military service members recently deployed to Afghanistan or Iraq, LINE-1 (long interspersed element, a DNA sequence that makes up 17% of the human genome) was hypermethylated among controls and hypomethylated postdeployment among participants with PTSD when compared with predeployment measurements (58). Alu, another repetitive DNA sequence, was also hypermethylated predeployment among participants who eventually were diagnosed with PTSD, when compared with controls (58). Thus, the methylation status of LINE-1 and Alu DNA may serve as a marker of exposure and susceptibility for service members for whom PTSD might occur, which suggests possible resilience or vulnerability factors. The SLC6A3 (solute carrier family 6 [neurotransmitter transporter, dopamine], member 3) locus has also been associated with increased risk of PTSD (59, 60). Other biomarkers that have been explored include inflammatory biomarkers, such as C-reactive protein (61, 62); variants of the gene-encoding FAAH (63, 64); the endocannabinoids anandamide and 2-AG (65, 66); arginine as a marker of nitric oxide synthesis capacity (67); telomere length (6870); and the interaction of attachment style and oxytocin gene polymorphisms (71).

Researchers have also worked to identify biomarkers by examining shared genetic characteristics across several disorders, through studies recruiting participants inclusive of PTSD, major depressive disorder, and anxiety disorders (72). Transdiagnostic investigation of G×E relationships may identify common biological underpinnings and allow for differentiation (73). This approach has also been used to examine core constructs within study samples that include nonclinical and subthreshold as well as clinical groups.

A goal of this approach is the identification of intermediate phenotypes or subtypes. Doing so may assist in the detection of significant loci in genomewide association studies (GWAS). GWAS studies for PTSD have proposed several loci, including rs406001 on 7p12 (74); PRTFDC1, a tumor suppressor gene highly expressed in the central nervous system (10); and ANKRD55 (75). In the largest PTSD GWAS thus far, with a sample size of N=20,730, no significant SNPs were identified, and results from prior studies were not replicated (76). The authors posited that much larger sample sizes are needed, along with the inclusion of more participants with no trauma exposure and improved phenotyping to decrease heterogeneity (76).

Clinical and Physiological Biomarkers

Researchers have investigated comorbidities and symptom clusters to further inform biological markers for PTSD. Examination of key clinical dimensions within PTSD—for example, specific symptoms, such as sleep disturbance (77)—has shown promise in further parsing the heterogeneity of PTSD populations. The most notable finding emerging from this approach has been the characterization of neurobiological findings related to PTSD with dissociative symptoms (78), which has resulted in the establishment of a dissociative subtype of PTSD in the DSM-5 (1, 79). Further extension of this set of findings resulted in a theoretical framework to distinguish PTSD symptoms associated with an altered state of consciousness from symptoms that are characteristic of a state of normal awareness (e.g., flashbacks vs. intrusive memories) (80, 81).

Clinical and physiological biomarkers, ones that are not blood-based bioassays, involve a broad set of approaches that includes several different types of measurement. Although the research use of psychophysiological measurements, such as skin conductance and heart rate variability (8288) and EEG (89, 90), in PTSD is longstanding, the use of such measures to characterize clinical subpopulations of PTSD for treatment selection is a promising area of current research. Exaggerated startle response is among the DSM-5 criteria for PTSD, which thus makes this symptom of particular relevance to study that uses psychophysiological methods, such as pupil response (91) or electromyography measurement of response in potentiated startle (92). Impaired inhibition of fear-potentiated startle has been studied among participants with PTSD, with the prospect for use of eye-blink reactivity as a differentiator of subtypes.

Neuroimaging data from functional magnetic resonance imaging and positron emission tomography studies have identified a number of brain regions that are likely involved in the pathophysiology of PTSD. These regions include the ventromedial PFC and anterior cingulate cortex, amygdala, hippocampus, and insula (22, 93). Although research has shown that these areas are affected among individuals with PTSD, the interplay among these regions may be more critical to the understanding and treatment of PTSD than are specific regional markers (94).

Increased amygdala activation in response to trauma-related stimuli supports the role of the amygdala in PTSD (22, 93). Studies of trauma-related stimuli have used script-driven imagery, aversive sounds, and backward-masked faces. Emotional content unrelated to trauma has also been examined in relation to increased amygdala activation among participants with PTSD compared with mentally healthy controls (93). Patients with PTSD also demonstrate increased activation of the amygdala at rest and in response to neutral stimuli, as well as greater activation when presented with fearful faces compared with neutral faces (93). Studies of the hippocampus in PTSD have shown mixed results, generally demonstrating possible decreased activation and decreased hippocampal volume among people with PTSD (22, 93).

The connectivity of regions involved in PTSD may be vital to understanding this complex disorder. The PFC, involved in executive function, serves a role in inhibition of the amygdala, a key locus for affective responses to trauma (22). The hippocampus, in parallel, further regulates the emotion-related activation of the overall system through memory and learning. In the context of a traumatic stimulus, the capacity of the PFC to provide control through inhibition is exceeded by increased activity in the amygdala and insula (93). Current efforts to combine functional neuroimaging with genomic data for enhanced phenotyping (94) and to use repeated measurements with machine learning to predict the time course of symptoms and recovery from PTSD (95) are examples of approaches that show promise for further clarifying neurobiological characteristics of PTSD.

Correlates of Treatment Response

The identification of phenotypic features of PTSD that are highly correlated with validated biomarker findings may result in clinically feasible subtyping and treatment selection. In the future, biomarkers could be used as part of an initial clinical evaluation to determine whether the patient is more likely to respond to medication or psychotherapy or to decide whether the best choice is a particular class of medications or one among several available psychotherapies. As discussed below, a number of studies have used biomarkers for identification of responder subgroups in treatment studies.

In a 2014 systematic review of structural and functional changes related to treatment effects, pharmacotherapy response was associated with improvement in structural abnormalities, whereas psychotherapy response was associated with decreased amygdala activation and increased activation of dorsolateral prefrontal regions (96). This suggests the possibility of detecting other biological predictors and correlates of response for specific treatments (see Box 2). Neuroimaging, however, is unlikely to be a preferred modality for biological measurement in the treatment setting, given feasibility and cost.

Fewer studies to date have examined biological markers of response to drug treatment for PTSD. In a small, open-label study of escitalopram, BDNF levels did not change significantly over the course of treatment. However, mean BDNF levels were correlated with the slope of symptom change—that is, lower mean BDNF was associated with a larger decrease in PTSD symptoms (97). In a secondary analysis of data from a randomized placebo-controlled trial of prazosin for PTSD, higher standing systolic blood pressure at baseline was associated with greater reductions in total Clinician-Administered PTSD Scale score (98), which suggests that blood pressure may be a biomarker for response to prazosin.

Research has more extensively focused on biomarkers of response to psychotherapy for PTSD. In a systematic review, neuroimaging studies generally showed that elevated activity in the dorsal anterior cingulate cortex was associated with treatment response, whereas elevated activity in the amygdala and insula at baseline were associated with lack of response. Successful therapy was associated with decreased activity in the amygdala and increased activation of the dorsal anterior cingulate cortex and hippocampus (99).

In a systematic review of baseline (pretreatment) biomarkers and psychotherapy outcomes in studies meeting Preferred Reporting Items for Systematic Reviews and Meta-Analyses criteria published prior to May 2016, brain regions and circuits implicated in processing trauma-related information and memory were predictive of response (100). In addition, markers related to the HPA axis, glucocorticoid sensitivity, and metabolism were predictive of response to psychotherapy. DNA methylation and genes related to glucocorticoids such as FKBP1 (101) and GR sensitivity, as well as elements in the serotonin system, were predictive of response (100). Increased baseline heart rate was also associated with response to psychotherapy. In a more recent report using psychophysiological measures and salivary cortisol at baseline prior to virtual-reality-based exposure therapy with and without d-cycloserine, increased reactivity to startle and lower cortisol reactivity to virtual reality were predictive of response (102).

Biomarkers—whether readily available ones, such as standing systolic blood pressure (as described above in one study of prazosin), or new tests that require a complex development process, such as a genetic assay—have promise for utility in the context of treatment selection and monitoring of response. Although biomarkers have been used to identify characteristics correlated with treatment response in clinical trials, the next step in furthering this approach involves the prospective use of a biomarker as an inclusion or exclusion criterion to define an enhanced study population with an increased likelihood of benefit. Doing so would serve several purposes to advance our understanding of PTSD therapeutics: Using a biomarker to select participants more likely to be well matched to the treatment might increase effect sizes, thereby allowing for improved signal detection. In addition, such biomarker use might improve treatment matching and early effectiveness when treatment is recommended for a particular individual.

Conclusion

In the context of advances in basic and translational research on biological characteristics of PTSD, it is challenging to extract relevant findings for practical application. The identification of reliable and accurate PTSD biomarkers will be a substantial breakthrough for improving clinical care. Ongoing awareness of advances in subtyping, clinical indicators of prognosis, and treatment matching can assist clinicians by providing context for individualized judgments.

Given that individual treatment needs are determined through a collaborative process between patient and clinician, the ultimate clinical utility of biomarkers as a part of precision medicine will enhance rather than supplant existing decision-making processes. The utility of biomarkers will be limited if the additional information from a biomarker does not exceed the clinician’s interview and any subtyping or treatment selection that can be established on the basis of phenomenology. Biomarkers have utility in research for identifying pathophysiological mechanisms and groups of individuals within the categorical diagnosis of PTSD. Attaining sufficient reliability and validity for use in individual treatment decisions is the hope of the near future.

The authors are with the Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University, Bethesda, Maryland. Dr. Yang is also with the Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland.
Send correspondence to Dr. Wynn (e-mail: ).

The views expressed are those of the authors and do not necessarily reflect the views of the Department of Defense, the Uniformed Services University, or the U.S. government.

The authors report no financial relationships with commercial interests.

References

1 Diagnostic and Statistical Manual of Mental Disorders, 5th ed. Arlington, VA, American Psychiatric Association, 2013Google Scholar

2 Galatzer-Levy IR, Bryant RA. 636,120 ways to have posttraumatic stress disorder. Perspect Psychol Sci 2013; 8:651-662.CrossrefGoogle Scholar

3 Drysdale AT, Grosenick L, Downar J, et al.: Resting-state connectivity biomarkers define neurophysiological subtypes of depression. Nat Med 2017; 23:28–38CrossrefGoogle Scholar

4 Dunlop BW, Rajendra JK, Craighead WE, et al.: Functional connectivity of the subcallosal cingulate cortex and differential outcomes to treatment with cognitive-behavioral therapy or antidepressant medication for major depressive disorder. Am J Psychiatry 2017; 174:533–545CrossrefGoogle Scholar

5 Ressler KJ, Schoomaker EB: Commentary on “The Army study to assess risk and resilience in servicemembers (Army STARRS)”: Army STARRS: a Framingham-like study of psychological health risk factors in soldiers. Psychiatry 2014; 77:120–129CrossrefGoogle Scholar

6 Yehuda R, Neylan TC, Flory JD, et al.: The use of biomarkers in the military: from theory to practice. Psychoneuroendocrinology 2013; 38:1912–1922CrossrefGoogle Scholar

7 Osuch E, Ursano R, Li H, et al.: Brain environment interactions: stress, posttraumatic stress disorder, and the need for a postmortem brain collection. Psychiatry 2004; 67:353–383CrossrefGoogle Scholar

8 National Posttraumatic Stress Disorder Brain Bank. Washington, DC, US Department of Veterans Affairs. https://www.research.va.gov/programs/tissue_banking/ptsd/default.cfm. Accessed Aug 14, 2017Google Scholar

9 Friedman MJ, Huber BR, Brady CB, et al.: VA’s National PTSD Brain Bank: a national resource for research. Curr Psychiatry Rep 2017; 19:73CrossrefGoogle Scholar

10 Nievergelt CM, Maihofer AX, Mustapic M, et al.: Genomic predictors of combat stress vulnerability and resilience in U.S. Marines: A genome-wide association study across multiple ancestries implicates PRTFDC1 as a potential PTSD gene. Psychoneuroendocrinology 2015; 51:459–471CrossrefGoogle Scholar

11 Almli LM, Stevens JS, Smith AK, et al.: A genome-wide identified risk variant for PTSD is a methylation quantitative trait locus and confers decreased cortical activation to fearful faces. Am J Med Genet B Neuropsychiatr Genet 2015; 168b:327–336CrossrefGoogle Scholar

12 Lehrner A, Yehuda R: Biomarkers of PTSD: military applications and considerations. Eur J Psychotraumatol 2014; 5: PMC4138702CrossrefGoogle Scholar

13 FDA-NIH Biomarker Working Group: BEST (biomarkers, endpoints, and other tools) resource. Silver Spring, MD: U.S. Food and Drug Administration and National Institutes of Health, 2016Google Scholar

14 Thakur GS, Daigle BJ Jr, Dean KR, et al.: Systems biology approach to understanding post-traumatic stress disorder. Mol Biosyst 2015; 11:980–993CrossrefGoogle Scholar

15 Galatzer-Levy IR, Karstoft KI, Statnikov A, et al.: Quantitative forecasting of PTSD from early trauma responses: a machine learning application. J Psychiatr Res 2014; 59:68–76CrossrefGoogle Scholar

16 Zhang L, Li H, Benedek D, et al.: A strategy for the development of biomarker tests for PTSD. Med Hypotheses 2009; 73:404–409CrossrefGoogle Scholar

17 Amur S, LaVange L, Zineh I, et al.: Biomarker qualification: toward a multiple stakeholder framework for biomarker development, regulatory acceptance, and utilization. Clin Pharmacol Ther 2015; 98:34–46CrossrefGoogle Scholar

18 Kraemer HC, Kazdin AE, Offord DR, et al.: Coming to terms with the terms of risk. Arch Gen Psychiatry 1997; 54:337–343CrossrefGoogle Scholar

19 Cabarkapa S, Perera M, McGrath S, et al.: Prostate cancer screening with prostate-specific antigen: a guide to the guidelines. Prostate Int 2016; 4:125–129CrossrefGoogle Scholar

20 US Preventive Services Task Force: Final Recommendation Statement. Prostate Cancer: Screening. https://www.uspreventiveservicestaskforce.org/Page/Document/RecommendationStatementFinal/prostate-cancer-screening. Accessed Aug 14, 2017Google Scholar

21 McLean SA: Longitudinal Assessment of Post-Traumatic Syndromes. Bethesda, MD, National Institute of Mental Health, 2016Google Scholar

22 Michopoulos V, Norrholm SD, Jovanovic T: Diagnostic biomarkers for posttraumatic stress disorder: promising horizons from translational neuroscience research. Biol Psychiatry 2015; 78:344–353CrossrefGoogle Scholar

23 Schmidt U, Willmund GD, Holsboer F, et al.: Searching for non-genetic molecular and imaging PTSD risk and resilience markers: systematic review of literature and design of the German Armed Forces PTSD biomarker study. Psychoneuroendocrinology 2015; 51:444–458CrossrefGoogle Scholar

24 Banerjee SB, Morrison FG, Ressler KJ: Genetic approaches for the study of PTSD: advances and challenges. Neurosci Lett 2017; 649:139–146CrossrefGoogle Scholar

25 Van Zuiden M, Kavelaars A, Geuze E, et al.: Predicting PTSD: pre-existing vulnerabilities in glucocorticoid-signaling and implications for preventive interventions. Brain Behav Immun 2013; 30:12–21CrossrefGoogle Scholar

26 Van Zuiden M, Geuze E, Willemen HL, et al.: Pre-existing high glucocorticoid receptor number predicting development of posttraumatic stress symptoms after military deployment. Am J Psychiatry 2011; 168:89–96CrossrefGoogle Scholar

27 Van Zuiden M, Geuze E, Willemen HL, et al.: Glucocorticoid receptor pathway components predict posttraumatic stress disorder symptom development: a prospective study. Biol Psychiatry 2012; 71:309–316CrossrefGoogle Scholar

28 Van Zuiden M, Heijnen CJ, Maas M, et al.: Glucocorticoid sensitivity of leukocytes predicts PTSD, depressive and fatigue symptoms after military deployment: a prospective study. Psychoneuroendocrinology 2012; 37:1822–1836CrossrefGoogle Scholar

29 Baker DG, Ekhator NN, Kasckow JW, et al.: Higher levels of basal serial CSF cortisol in combat veterans with posttraumatic stress disorder. Am J Psychiatry 2005; 162:992–994CrossrefGoogle Scholar

30 Bremner JD, Licinio J, Darnell A, et al.: Elevated CSF corticotropin-releasing factor concentrations in posttraumatic stress disorder. Am J Psychiatry 1997; 154:624–629CrossrefGoogle Scholar

31 Geracioti TD Jr, Baker DG, Kasckow JW, et al.: Effects of trauma-related audiovisual stimulation on cerebrospinal fluid norepinephrine and corticotropin-releasing hormone concentrations in post-traumatic stress disorder. Psychoneuroendocrinology 2008; 33:416–424CrossrefGoogle Scholar

32 White S, Acierno R, Ruggiero KJ, et al.: Association of CRHR1 variants and posttraumatic stress symptoms in hurricane exposed adults. J Anxiety Disord 2013; 27:678–683CrossrefGoogle Scholar

33 Yehuda R, Flory JD, Bierer LM, et al.: Lower methylation of glucocorticoid receptor gene promoter 1F in peripheral blood of veterans with posttraumatic stress disorder. Biol Psychiatry 2015; 77:356–364CrossrefGoogle Scholar

34 Zannas AS, Binder EB: Gene-environment interactions at the FKBP5 locus: sensitive periods, mechanisms and pleiotropism. Genes Brain Behav 2014; 13:25–37CrossrefGoogle Scholar

35 Gillespie CF, Phifer J, Bradley B, et al.: Risk and resilience: genetic and environmental influences on development of the stress response. Depress Anxiety 2009; 26:984–992CrossrefGoogle Scholar

36 Binder EB, Bradley RG, Liu W, et al.: Association of FKBP5 polymorphisms and childhood abuse with risk of posttraumatic stress disorder symptoms in adults. JAMA 2008; 299:1291–1305CrossrefGoogle Scholar

37 Watkins LE, Han S, Harpaz-Rotem I, et al.: FKBP5 polymorphisms, childhood abuse, and PTSD symptoms: results from the National Health and Resilience in Veterans Study. Psychoneuroendocrinology 2016; 69:98–105CrossrefGoogle Scholar

38 Mehta D, Gonik M, Klengel T, et al.: Using polymorphisms in FKBP5 to define biologically distinct subtypes of posttraumatic stress disorder: evidence from endocrine and gene expression studies. Arch Gen Psychiatry 2011; 68:901–910CrossrefGoogle Scholar

39 Klengel T, Mehta D, Anacker C, et al.: Allele-specific FKBP5 DNA demethylation mediates gene-childhood trauma interactions. Nat Neurosci 2013; 16:33–41CrossrefGoogle Scholar

40 Hammack SE, May V: Pituitary adenylate cyclase activating polypeptide in stress-related disorders: data convergence from animal and human studies. Biol Psychiatry 2015; 78:167–177CrossrefGoogle Scholar

41 Dias BG, Ressler KJ. PACAP and the PAC1 receptor in post-traumatic stress disorder. Neuropsychopharmacology 2013; 38:245-246.CrossrefGoogle Scholar

42 Licznerski P, Duric V, Banasr M, et al.: Decreased SGK1 expression and function contributes to behavioral deficits induced by traumatic stress. PLoS Biol 2015; 13:e1002282CrossrefGoogle Scholar

43 Zhang L, Li H, Su TP, et al.: p11 is up-regulated in the forebrain of stressed rats by glucocorticoid acting via two specific glucocorticoid response elements in the p11 promoter. Neuroscience 2008; 153:1126–1134CrossrefGoogle Scholar

44 Zhang L, Li H, Hu X, et al.: Glucocorticoid-induced p11 over-expression and chromatin remodeling: a novel molecular mechanism of traumatic stress? Med Hypotheses 2011; 76:774–777CrossrefGoogle Scholar

45 Su TP, Zhang L, Chung MY, et al.: Levels of the potential biomarker p11 in peripheral blood cells distinguish patients with PTSD from those with other major psychiatric disorders. J Psychiatr Res 2009; 43:1078–1085CrossrefGoogle Scholar

46 Andero R, Choi DC, Ressler KJ: BDNF-TrkB receptor regulation of distributed adult neural plasticity, memory formation, and psychiatric disorders. Prog Mol Biol Transl Sci 2014; 122:169–192CrossrefGoogle Scholar

47 Bennett MR, Lagopoulos J: Stress and trauma: BDNF control of dendritic-spine formation and regression. Prog Neurobiol 2014; 112:80–99CrossrefGoogle Scholar

48 Benarroch EE: Brain-derived neurotrophic factor: regulation, effects, and potential clinical relevance. Neurology 2015; 84:1693–1704CrossrefGoogle Scholar

49 Van den Heuvel L, Suliman S, Malan-Müller S, et al.: Brain-derived neurotrophic factor Val66met polymorphism and plasma levels in road traffic accident survivors. Anxiety Stress Coping 2016; 29:616–629CrossrefGoogle Scholar

50 Dell’Osso L, Carmassi C, Del Debbio A, et al.: Brain-derived neurotrophic factor plasma levels in patients suffering from post-traumatic stress disorder. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:899–902CrossrefGoogle Scholar

51 Hauck S, Kapczinski F, Roesler R, et al.: Serum brain-derived neurotrophic factor in patients with trauma psychopathology. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34:459–462CrossrefGoogle Scholar

52 Zhang L, Benedek DM, Fullerton CS, et al.: PTSD risk is associated with BDNF Val66Met and BDNF overexpression. Mol Psychiatry 2014; 19:8–10CrossrefGoogle Scholar

53 Zhang H, Ozbay F, Lappalainen J, et al.: Brain derived neurotrophic factor (BDNF) gene variants and Alzheimer's disease, affective disorders, posttraumatic stress disorder, schizophrenia, and substance dependence. Am J Med Genet B Neuropsychiatr Genet 2006; 141B:387-393.CrossrefGoogle Scholar

54 Valente NL, Vallada H, Cordeiro Q, et al.: Candidate-gene approach in posttraumatic stress disorder after urban violence: association analysis of the genes encoding serotonin transporter, dopamine transporter, and BDNF. J Mol Neurosci 2011; 44:59-67.CrossrefGoogle Scholar

55 Klengel T, Pape J, Binder EB, et al.: The role of DNA methylation in stress-related psychiatric disorders. Neuropharmacology 2014; 80:115–132CrossrefGoogle Scholar

56 Zannas AS, Provençal N, Binder EB: Epigenetics of posttraumatic stress disorder: current evidence, challenges, and future directions. Biol Psychiatry 2015; 78:327–335CrossrefGoogle Scholar

57 Xie P, Kranzler HR, Farrer L, et al.: Serotonin transporter 5-HTTLPR genotype moderates the effects of childhood adversity on posttraumatic stress disorder risk: a replication study. Am J Med Genet B Neuropsychiatr Genet 2012; 159b:644-652.CrossrefGoogle Scholar

58 Rusiecki JA, Chen L, Srikantan V, et al.: DNA methylation in repetitive elements and post-traumatic stress disorder: a case-control study of US military service members. Epigenomics 2012; 4:29–40CrossrefGoogle Scholar

59 Li L, Bao Y, He S, et al.: The association between genetic variants in the dopaminergic system and posttraumatic stress disorder: a meta-analysis. Medicine 2016; 95:e3074CrossrefGoogle Scholar

60 Chang SC, Koenen KC, Galea S, et al.: Molecular variation at the SLC6A3 locus predicts lifetime risk of PTSD in the Detroit Neighborhood Health Study. PLoS One 2012; 7:e39184CrossrefGoogle Scholar

61 Eraly SA, Nievergelt CM, Maihofer AX, et al.: Assessment of plasma C-reactive protein as a biomarker of posttraumatic stress disorder risk. JAMA Psychiatry 2014; 71:423–431CrossrefGoogle Scholar

62 Michopoulos V, Powers A, Gillespie CF, et al.: Inflammation in fear- and anxiety-based disorders: PTSD, GAD, and beyond. Neuropsychopharmacology 2017; 42:254-270.CrossrefGoogle Scholar

63 Spagnolo PA, Ramchandani VA, Schwandt ML, et al.: FAAH gene variation moderates stress response and symptom severity in patients with posttraumatic stress disorder and comorbid alcohol dependence. Alcohol Clin Exp Res 2016; 40:2426–2434CrossrefGoogle Scholar

64 Pardini M, Krueger F, Koenigs M, et al.: Fatty-acid amide hydrolase polymorphisms and post-traumatic stress disorder after penetrating brain injury. Transl Psychiatry 2012; 2:e75CrossrefGoogle Scholar

65 Hill MN, Bierer LM, Makotkine I, et al.: Reductions in circulating endocannabinoid levels in individuals with post-traumatic stress disorder following exposure to the World Trade Center attacks. Psychoneuroendocrinology 2013; 38:2952–2961CrossrefGoogle Scholar

66 Neumeister A, Normandin MD, Pietrzak RH, et al.: Elevated brain cannabinoid CB1 receptor availability in post-traumatic stress disorder: a positron emission tomography study. Mol Psychiatry 2013; 18:1034–1040CrossrefGoogle Scholar

67 Bersani FS, Wolkowitz OM, Lindqvist D, et al.: Global arginine bioavailability, a marker of nitric oxide synthetic capacity, is decreased in PTSD and correlated with symptom severity and markers of inflammation. Brain Behav Immun 2016; 52:153–160CrossrefGoogle Scholar

68 Lohr JB, Palmer BW, Eidt CA, et al.: Is post-traumatic stress disorder associated with premature senescence? A review of the literature. Am J Geriatr Psychiatry 2015; 23:709–725CrossrefGoogle Scholar

69 Zhang L, Hu XZ, Li X, et al.: Telomere length—a cellular aging marker for depression and post-traumatic stress disorder. Med Hypotheses 2014; 83:182–185CrossrefGoogle Scholar

70 Zhang L, Hu XZ, Benedek DM, et al.: The interaction between stressful life events and leukocyte telomere length is associated with PTSD. Mol Psychiatry 2014; 19:855–856CrossrefGoogle Scholar

71 Sippel LM, Han S, Watkins LE, et al.: Oxytocin receptor gene polymorphisms, attachment, and PTSD: results from the National Health and Resilience in Veterans Study. J Psychiatr Res 2017; 94:139–147CrossrefGoogle Scholar

72 Smoller JW. The genetics of stress-related disorders: PTSD, depression, and anxiety disorders. Neuropsychopharmacology 2016; 41:297-319.CrossrefGoogle Scholar

73 Montalvo-Ortiz JL, Gelernter J, Hudziak J, Kaufman J. RDoC and translational perspectives on the genetics of trauma-related psychiatric disorders. Am J Med Genet B Neuropsychiatr Genet 2016; 171b:81-91.CrossrefGoogle Scholar

74 Xie P, Kranzler HR, Yang C, et al: Genome-wide association study identifies new susceptibility loci for posttraumatic stress disorder. Biol Psychiatry 2013; 74:656–663Google Scholar

75 Stein MB, Chen CY, Ursano RJ, et al: Genome-wide association studies of posttraumatic stress disorder in 2 cohorts of US Army soldiers. JAMA Psychiatry 2016; 73:695–704Google Scholar

76 Duncan LE, Ratanatharathorn A, Aiello AE, et al.: Largest GWAS of PTSD (N=20 070) yields genetic overlap with schizophrenia and sex differences in heritability. Mol Psychiatry (Epub ahead of print, Apr 25, 2017)Google Scholar

77 Krystal JH, Pietrzak RH, Rosenheck RA, et al.: Sleep disturbance in chronic military-related PTSD: clinical impact and response to adjunctive risperidone in the Veterans Affairs cooperative study #504. J Clin Psychiatry 2016; 77:483–491CrossrefGoogle Scholar

78 Lanius RA, Brand B, Vermetten E, et al.: The dissociative subtype of posttraumatic stress disorder: rationale, clinical and neurobiological evidence, and implications. Depress Anxiety 2012; 29:701–708CrossrefGoogle Scholar

79 Spiegel D, Lewis-Fernández R, Lanius R, et al.: Dissociative disorders in DSM-5. Annu Rev Clin Psychol 2013; 9:299–326CrossrefGoogle Scholar

80 Lanius RA: Trauma-related dissociation and altered states of consciousness: a call for clinical, treatment, and neuroscience research. Eur J Psychotraumatol 2015; 6:27905CrossrefGoogle Scholar

81 Frewen PA, Lanius RA. Trauma-related altered states of consciousness: exploring the 4-D model. J Trauma Dissociation 2014;15:436-456CrossrefGoogle Scholar

82 Agorastos A, Boel JA, Heppner PS, et al.: Diminished vagal activity and blunted diurnal variation of heart rate dynamics in posttraumatic stress disorder. Stress 2013; 16:300–310CrossrefGoogle Scholar

83 Dennis PA, Dedert EA, Van Voorhees EE, et al.: Examining the crux of autonomic dysfunction in posttraumatic stress disorder: whether chronic or situational distress underlies elevated heart rate and attenuated heart rate variability. Psychosom Med 2016; 78:805–809CrossrefGoogle Scholar

84 Minassian A, Maihofer AX, Baker DG, et al.: Association of predeployment heart rate variability with risk of postdeployment posttraumatic stress disorder in active-duty Marines. JAMA Psychiatry 2015; 72:979–986CrossrefGoogle Scholar

85 Minassian A, Geyer MA, Baker DG, et al.: Heart rate variability characteristics in a large group of active-duty Marines and relationship to posttraumatic stress. Psychosom Med 2014; 76:292–301CrossrefGoogle Scholar

86 Pyne JM, Constans JI, Wiederhold MD, et al.: Heart rate variability: pre-deployment predictor of post-deployment PTSD symptoms. Biological Psychology 2016; 121(Pt A):91-9ß8.CrossrefGoogle Scholar

87 Rabellino D, D’Andrea W, Siegle G, et al.: Neural correlates of heart rate variability in PTSD during sub- and supraliminal processing of trauma-related cues. Hum Brain Mapp 2017; 38:4898–4907CrossrefGoogle Scholar

88 Rissling MB, Dennis PA, Watkins LL, et al.: Circadian contrasts in heart rate variability associated with posttraumatic stress disorder symptoms in a young adult cohort. J Trauma Stress 2016; 29:415–421CrossrefGoogle Scholar

89 Lobo I, Portugal LC, Figueira I, et al.: EEG correlates of the severity of posttraumatic stress symptoms: a systematic review of the dimensional PTSD literature. J Affect Disord 2015; 183:210–220CrossrefGoogle Scholar

90 Meyer T, Smeets T, Giesbrecht T, et al.: The role of frontal EEG asymmetry in post-traumatic stress disorder. Biol Psychol 2015; 108:62–77CrossrefGoogle Scholar

91 Cascardi M, Armstrong D, Chung L, et al.: Pupil response to threat in trauma-exposed individuals with or without PTSD. J Trauma Stress 2015; 28:370–374CrossrefGoogle Scholar

92 McTeague LM, Lang PJ, Laplante MC, et al.: Aversive imagery in posttraumatic stress disorder: trauma recurrence, comorbidity, and physiological reactivity. Biol Psychiatry 2010; 67:346–356CrossrefGoogle Scholar

93 Hughes KC, Shin LM: Functional neuroimaging studies of post-traumatic stress disorder. Expert Rev Neurother 2011; 11:275–285CrossrefGoogle Scholar

94 Miller DR, Logue MW, Wolf EJ, et al.: Posttraumatic stress disorder symptom severity is associated with reduced default mode network connectivity in individuals with elevated genetic risk for psychopathology. Depress Anxiety 2017; 34:632–640CrossrefGoogle Scholar

95 Im JJ, Kim B, Hwang J, et al.: Diagnostic potential of multimodal neuroimaging in posttraumatic stress disorder. PLoS One 2017; 12:e0177847CrossrefGoogle Scholar

96 Thomaes K, Dorrepaal E, Draijer N, et al.: Can pharmacological and psychological treatment change brain structure and function in PTSD? A systematic review. J Psychiatr Res 2014; 50:1–15CrossrefGoogle Scholar

97 Berger W, Mehra A, Lenoci M, et al.: Serum brain-derived neurotrophic factor predicts responses to escitalopram in chronic posttraumatic stress disorder. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34:1279–1284CrossrefGoogle Scholar

98 Raskind MA, Millard SP, Petrie EC, et al.: Higher pretreatment blood pressure is associated with greater posttraumatic stress disorder symptom reduction in soldiers treated with prazosin. Biol Psychiatry 2016; 80:736–742CrossrefGoogle Scholar

99 Malejko K, Abler B, Plener PL, et al.: Neural correlates of psychotherapeutic treatment of post-traumatic stress disorder: a systematic literature review. Front Psychiatry 2017; 8:85CrossrefGoogle Scholar

100 Colvonen PJ, Glassman LH, Crocker LD, et al.: Pretreatment biomarkers predicting PTSD psychotherapy outcomes: a systematic review. Neurosci Biobehav Rev 2017; 75:140–156CrossrefGoogle Scholar

101 Yehuda R, Daskalakis NP, Desarnaud F, et al.: Epigenetic biomarkers as predictors and correlates of symptom improvement following psychotherapy in combat veterans with PTSD. Front Psychiatry 2013; 4:118CrossrefGoogle Scholar

102 Norrholm SD, Jovanovic T, Gerardi M, et al.: Baseline psychophysiological and cortisol reactivity as a predictor of PTSD treatment outcome in virtual reality exposure therapy. Behav Res Ther 2016; 82:28–37CrossrefGoogle Scholar