Loading

JSM Biomarkers

How can we Track Psychosis? A Scoping Review of Biomarkers of Transition in Subjects at Risk

Review Article | Open Access | Volume 5 | Issue 1

  • 1. Centre de recherche CERVO, Québec City, QC, Canada
  • 2. Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Canada
  • 3. Laboratoire Santé mentale des jeunes et technologies (SMJ-techno), Canada
  • 4. Département de psychiatrie et neurosciences, Université Laval, Canada
+ Show More - Show Less
Corresponding Authors
Alexandra Painchaud, Centre de recherche CERVO, 2601 chemin de la Canardière, Québec City, QC, Canada, Tel: 1 (581) 703-3838; Email : alexandra.painchaud.2@ulaval.ca
ABSTRACT

In recent years, research has made progress in attempting to foresee the development of psychosis. Several definitions of clinical syndromes and/or familial susceptibility have been proposed to predict transition to psychosis. Among subjects identified as being at risk, it is expected that almost a third of them will eventually transition to psychosis. Although these definitions clearly represent sensitive tools, they lack specificity and rely on clinical judgment. Hence, biomarkers are being investigated as objective biological assessment tools to refine the prediction of psychosis. This scoping review focuses on the biomarkers currently under investigation. We reviewed the studies published since January 2015 that put forth a biomarker associated with the risk of developing schizophrenia (SZ) or a SZ spectrum disorder. A search of the MEDLINE (via PubMed), EMBASE, PsycINFO (via OVID) and Cochrane Central Register of Controlled Trials electronic databases was conducted and 96 studies that investigated predictive biomarkers in the at-risk population were included. More precisely, we reviewed these studies focusing on study design, the effect size of the association between biomarkers and psychosis, and evidence for replication. The review revealed several biomarkers, including reduced duration mismatch negativity, reduced 40 Hz auditory steady-state responses and impaired gyrification parameters. In this paper, we discuss the lack of clinical use of these potential tools and the criteria that biomarkers must meet to be considered as medical guidelines in psychiatry.

KEYWORDS

Psychosis; biomarkers; schizophrenia; high risk; transition.

CITATION

Painchaud A, Peredo R, Mérette C, Marquet P (2022) How can we Track Psychosis? A Scoping Review of Biomarkers of Transition in Subjects at Risk. JSM Biomar 5(1): 1014.

DISCUSSION

Since psychiatric diagnoses are mainly based on clinical observations and interviews, objective tests like biomarkers could be helpful to limit the duration of untreated prodromal symptoms [96]. Our main goal was to scope the literature to pin down biomarkers predictive of or associated with psychosis currently under study as reported since January 2015. We have mapped over 80 biomarkers which could have the potential to track early signs of SZ, which varied from brain imaging and genetics to proteins and retinal abnormalities.

We paid particular attention to the longitudinal studies that predicted transition of first-episode psychosis and to the effect sizes that characterized correlations between biomarkers and psychosis. Sample size, the length of longitudinal follow-ups and evidence of replication were also considered as important characteristics to highlight biomarkers. According to these criteria, a few studies stood out

Thanks to a longitudinal study design, Koike et al. [31] found that frontal waveforms (fNIRS assessment) predicted 100% of the transition to psychosis of six particular REP subjects. This result could be the first evidence of the correlation between frontal waveforms and risks of transition, although previous studies had already related this biomarker to chronic patients [97]. Despite the small sample size used and the need for replication, fNIRS assessment offers the benefits of a small, non-invasive and low-noise instrument that is easy to carry around compared with other brain-imaging techniques. Similarly, salivary cortisol is a non-invasive biomarker that showed a 21.5-fold risk of transitioning to psychosis in a cohort of 417 REP subjects [37]; in fact, cortisol abnormalities had already been seen in SZ patients [98,99]. However, the study of hormonal changes represents a challenge, given the high inter-individual variability. In addition, auditory P300 event-related potential has been studied since the 1980s as a primary electrophysiological biomarker candidate for psychosis [100]. More recently, Higuchi et al. [24] and Hamilton et al. [27,28] contributed to enhancing the potential of P300 event-related responses or its subcomponent, the P3b, as biomarkers of transition to psychosis in 8, 73 and 15 REP-T subjects, respectively. Transition to psychosis was also predicted by reduced 40 Hz auditory steady-state responses (ASSRs) in 116 REP subjects among which 13 transitioned [30]. Reduced 40 Hz ASSRs were also observed in REP subjects compared with control subjects during two other studies assessed in this review [68,69].

Other biomarkers were found to clearly distinguish REP-T and REP-NT subjects. Higher brain transitivity was found to be strongly related to transition to psychosis when 16 subjects who transitioned were compared with 63 others who did not (Hedges g = 1.76) and with 44 control subjects (Hedges g = 1.41) [14]. Although studies before 2015 had already investigated brain networks in SZ patients [101–103] the Das et al. study [14] seems to be the first to relate brain transitivity to transition to psychosis. Similarly, decreased microstate D strongly differed between 20 REP-T and 34 REP-NT subjects (Cohen’s d = -1.24) [25]. Bock et al. [25] were the first to assess microstates with respect to future transition to psychosis. Furthermore, the longitudinal decreased duration mismatch negativity (dMMN) amplitudes found in 11 REP-T subjects [26] seem promising. Since 2015, four other studies found decreased dMMN in REP subjects compared with control subjects [64-67].

Since 2015, no other clear replications of biomarkers to distinguish REP-T and REP-NT subjects were observed. However, some studies shared a similar purpose. For instance, subjects who transitioned had hypergyrification, i.e. higher levels of local gyrification index (LGI) in the left occipital region, compared with REP-NT subjects [20]. In contrast, other studies that compared subjects at risk with control subjects found hypogyrification (lower levels of LGI) in the lateral orbitofrontal, superior bank of the superior temporal sulcus, anterior isthmus of the cingulate and temporal poles [39] and in the medial parietooccipital and cingulate regions [40]. These opposite results could be explained by differences in the brain regions studied and the use of REP subjects disregarding who had transitioned. Previous investigations of gyrification in REP subjects suggested hypergyrification of frontal and parietal regions [104,105] with frontal hypergyrification associated with later development of a SZ spectrum disorder [106]. Gyrification parameters thus seem a promising avenue to predict transition.

As an alternative to follow-up studies, cluster analysis was proposed to identify subgroups of REP subjects according to a biomarker assessment (e.g., Peredo et al. [90]).The subgroups identified were then related to cognitive functioning, with the hypothesis that a low-functioning cluster would include subjects who will eventually transition to psychosis. Likewise, nine other studies have stratified REP subjects into subjects with high (REP+ ) versus low deficits (REP- ; Table 3), in order to identify a biomarker related to the REP+ state, assuming it would predict transition.

Most of the 96 studies assessed in this review had related a biomarker of interest to the state of being at risk compared with being a control subject, but not with transition to psychosis itself (78 studies in Table 3). Although such biomarkers still need to be further investigated in longitudinal design targeting REP-T subjects, these studies provided biomarker candidates, including mismatch negativity, 40 Hz ASSRs and P300 peak amplitude. These potential biomarkers have shown consistent differences between REP and control subjects in at least two independent cohorts assessed since 2015. However, there was a lack of consistency across studies assessing the same biomarker due to the wide range of instruments and measurements used.

CLINICAL USE

Early treatment is known to improve the prognosis of SZ spectrum disorders by reducing the severity of symptoms and improving long-term functioning [107–109]. Hence, in the last version of the Diagnostic and Statistical Manual of Mental Disorders (DSM-5, 2013), researchers attempted to add biomarkers as external validations to enhance diagnoses [110]. This idea was apparently premature, as biomarkers are not featured in DSM-5 [18]. Still, the keen interest in biomarkers in the last decade has stimulated research on biological assessments likely to monitor the development of a psychiatric disorder in the same way that mammography serves to monitor breast cancer [111]. Reliable, reproducible and non-invasive biomarkers would help to inform clinical guidelines on how and when to provide care to subjects slipping into psychosis. Several biomarkers are currently under study for this very purpose. From this perspective, biomarkers could be useful in walk-in clinics [112] where, for instance, young people seek various health and wellness services, before diagnosis of psychiatric illness. However, further research needs to better understand the stages in the progression of psychotic disorders, in order for biomarkers to be adequately integrated into follow-up processes provided to young individuals in need of care

LIMITATIONS

We limited the scope of our review to studies conducted since January 2015 to identify biomarkers being currently investigated, although it cannot be excluded that an earlier study could eventually provide a relevant biomarker. In this review, we also included studies with variegated definitions of REP subjects (genetic and/or clinical susceptibilities). Assessing genetic and clinical risks combined can be unadvisable, considering that they are not based on the same concept of risk. Our review also highlighted the difficulty of clearly identifying an individual’s age at which a given biomarker was assessed and, hence, if it truly revealed major neurodevelopmental changes in the individual’s brain during the lead time to the onset of psychosis. Lastly, we limited this review to assessing the risk of SZ spectrum disorders, although first-episode psychosis could also lead to bipolar disorder [113,114].

CONCLUSION

Several parameters need to be considered to assess the relevance of a biomarker, including: ease of use, cost/benefit ratio, level of intrusiveness, accuracy of prognostics, and replication and consistency across studies. No biomarker of psychosis has clearly met all these criteria as of yet, but research is in progress. By this review, we aim to inform practitioners in the field that several promising candidates have been identified in order to eventually be able to track psychosis and SZ spectrum disorders. Also, given that a number of causal mechanisms for psychiatric disorders have been acknowledged [115] and that heterogeneity and comorbidity are part of the portrait, several biomarkers may be needed and, perhaps, combined to determine the development of psychosis in individuals. Moreover, all biomarkers currently under investigation can be promising in a specific developmental stage. While further research still needs to assess the when and where of a biomarker, studies in the field are promising and the translational clinical benefits are about to be revealed.

DECLARATION OF COMPETING INTERESTS

The authors hereby declare that they have no known competing financial interests or personal relationships that could appear to have influenced the work reported in this paper.

CONFLICTS OF INTEREST

C. Mérette, Professor at Université Laval is listed as co-inventor in a patent application (Appl. No.: 16/685960) entitled “Use of electroretinography (ERG) for the assessment of psychiatric disorders” and holds shares in a start-up company (diaMentis), which owns a license from Université Laval to further develop and market the claims listed in the patent application. A. Painchaud reported no biomedical financial interests or potential conflicts of interest. R. Peredo Nunez De Arco reported no biomedical financial interests or potential conflicts of interest. P. Marquet reported no biomedical financial interests or potential conflicts of interest.

ACKNOWLEDGEMENTS

The authors would like to acknowledge the support of the Canadian Institutes of Health Research funding bodies

ABBREVIATIONS

SZ: Schizophrenia; SMI: Severe Mental Illness; HR: High-risk subjects; FHR: Familial “genetic” high-risk; CHR: Clinical High Risk; ARMS: At-risk mental state; UHR: Ultra-high risk; PRS: Psychotic Risk Syndrome; REP: Subjects at Risk of first-Episode Psychosis; REP-T: REP subjects who later Transitioned to psychosis; REP-NT: REP subjects who did Not Transition; sMRI: Structural Magnetic Resonance Imaging; LGI: Local Gyrification Index; GMV: Gray Matter Volume; ACG: Anterior Cingulate Gyrus; EEG: Electroencephalogram; dMMN: Duration Mismatch Negativity; fMRI: Functional Magnetic Resonance Imaging; rs-fMRI: Resting state functional Magnetic Resonance Imaging; ASSR: Auditory steady- state responses; MEG: Magnetoencephalography; MRS: Magnetic Resonance Spectroscopy; fNIRS: Functional nearinfrared Spectroscopy; PET: Position Emission Tomography; EMG: Electromyography; NS: Niacin Skin Sensitivity; REP+ : REP subjects with a higher level of deficits; REP- : REP subjects with a lower level of deficits; FEP: Subjects with first-episode psychosis; PFC: Prefrontal Cortex; IPL: Inferior Parietal Lobule; PCC: Posterior Cingulate Cortex; FCS: Functional Connectivity Strength; ReHo: Regional Homogeneity; ITG: Inferior Temporal Gyrus; DTI: Diffusion Tensor Imaging; DWI: Diffusion-weighted Imaging; TSPO: Translocator Protein; ERG: Electroretinography; HRV: Heart Rate Variability

INTRODUCTION

Schizophrenia (SZ) spectrum disorders are mostly chronic mental disorders, characterized by psychotic, positive or negative symptoms and cognitive deficits [1,2]. The full extent of these diseases usually strikes in early adulthood or adolescence [3]. However, a scientific consensus says that neurobiological alterations emerge even before the first episode of psychosis [4-6], suggesting a neurodevelopmental component [7-9] likely to be measured as a predictive biomarker in the population at risk. Hence, finding indicators of the biological processes underlying the neurodevelopmental trajectories of SZ could offer the opportunity to track early signs of mental illness in order to prevent or delay transition to psychosis

Having a biological parent with a severe mental illness (SMI) – such as SZ– is known to be a psychosis risk factor among offspring referred to as high-risk subjects (HR; or familial “genetic” highrisk; FHR) [10]. In addition to this genetic susceptibility, clinical symptoms like subthreshold or attenuated psychotic symptoms and impairment in day-to-day functioning have also been identified as psychosis risk factors [11]. In recent literature, subjects at clinical risk for psychosis are referred to as subjects with a clinical high risk (CHR), an at-risk mental state (ARMS), an ultra-high risk (UHR), a psychotic risk syndrome (PRS), or an attenuated psychosis syndrome (see Table 1 for definitions and

Table 1: Definitions of subjects at Risk of first-Episode Psychosis (REP) found in current literature are provided below.

 

REP Definition

Criteria

 

Genetic risk

High-risk (HR) or familial

 

First-degree familial history of SZ spectrum disorder [67]

high-risk (FHR)

 

Clinical risk

 

Clinical high risk (CHR)

  1. Diagnostic criteria for prodromal syndrome
  2. No current DSM Axis I disorder
  3. No history of or current treatment with antipsychotic medication [76]

 

 

At-risk mental state (ARMS)

  1. No previous episode of overt psychosis

+ One or more:

  1. Attenuated psychotic symptoms
  2. Brief and limited intermittent psychotic symptoms
  3. Genetic risk
  4. Deterioration syndrome [44]

 

Ultra-high risk (UHR)

One or more:

  1. Attenuated positive symptoms
  2. Brief limited intermittent psychotic symptoms
  3. Mental state and trait risk factors with functional decline [116]

 

 

Psychotic risk syndrome (PRS)

One or more:

  1. Attenuated psychotic symptoms
  2. Brief limited intermittent psychotic symptoms
  3. Familial history of psychosis in first-degree relatives
  4. Change in functioning occurred within the last year and maintained at least one month [11]

references). For the purpose of this review, subjects falling into any of these genetic and/or clinical susceptibility categories will be referred to overall as subjects at risk of first- psychosis (REP). As much as 30% of REP subjects will eventually develop an SMI in adulthood, especially after experiencing a first episode of psychosis [12, 13]. It remains that most of them will not develop a SZ spectrum disorder. Hence, based solely on this classification (REP), it is impossible to make any prediction on the onset of individual psychosis [14]. Furthermore, early intervention trials among REP subjects to delay or prevent the onset of psychosis (e.g., cognitive behavioral therapy, omega-3 fatty acids, or integrated psychotherapy) have shown moderate efficiency [15]. This finding could be partly due to the difficulty of specifically treating subjects slipping into psychosis. Thus, clinical trials would benefit from an enhanced capacity to identify, among REP subjects, those who are more likely to develop psychosis. Given the lack of specificity in the REP classification, biomarkers are needed to better identify the minority of individuals who will slip into psychosis.

Biomarkers are objective indicators of biological processes [16] that are already implemented in preventive medicine. Although research on biomarkers in the field of psychiatry has made significant progress since the beginning of the new millennium 2000s [16-18], there is yet to be a translational use in the clinical setting. Why? What is the current state of research on psychosis biomarkers? By reviewing studies published since 2015, we have mapped the available evidence in order to scope predictive biomarkers in REP subjects. We highlighted the study design used, the effect size of the correlation between biomarkers and psychosis, and the evidence for replication. In this paper, we will discuss the lack of clinical psychiatry tools and we will attempt to identify potential biomarkers for future clinical use.

METHOD

We have decided to do a scoping review [19] to cover the body of recent literature on biomarkers associated with the risk of developing a SZ-related disorder. We searched for papers on studies that investigated predictive biomarkers measured in REP subjects. We used the MEDLINE (via PubMed), EMBASE, PsycINFO (via OVID), and Cochrane Central Register of Controlled Trials electronic databases to search for a combination of indexing terms and free-text words in titles or abstracts referring to our population of interest: “Clinical high risk state for psychosis” OR “High risk state for psychosis” OR “Ultra High risk for psychosis” OR “Clinical high risk for psychosis” OR “Clinical risk for psychosis” OR "at-risk mental state for psychosis” OR “risk for psychosis” OR “psychosis risk” OR “psychosis high risk” OR “psychosis prediction” OR “psychosis prodrome” OR “prodromal psychosis” OR “psychosis*” OR “psychotic” OR “psychotic disorder” OR “psychotic episode” OR “early psychosis” OR “first episode” OR “schizophrenia” OR “schizoaffective” OR “Schizophrenia Spectrum and Other Psychotic Disorders”, combined with the terms “biomarker” OR “marker”. This review included English-language papers only published since January 2015, excluding conference abstracts, editorials, book chapters, animal studies, reviews, and meta-analyses.

As shown in the flowchart (Figure 1),

 Flowchart of Selection Process.

Figure 1 :Flowchart of Selection Process.

the search yielded 2,945 potential studies after duplicates were removed. Of those, 200 studies met inclusion criteria (predictive biomarkers measured in REP subjects) after abstracts were screened, and 96 studies remained after the full texts were reviewed.

RESULTS

Ninety-six articles met our inclusion criteria (Figure 1). The results from the 19 studies addressing transition to psychosis (Section 1) will be presented first, followed by the results from the studies on the state of being a REP subject without observing transition (Section 2). Each section will then be subdivided according to the type of biomarker (brain, blood, genetic, eye, or others), study design, and group comparison.

Psychosis transition biomarkers

Given that not all REP subjects will eventually transition to psychosis, our main interest was to review the tools (biomarkers) likely to predict the subjects who will transition. Follow-up studies allow for the assessment of such transition. Table 2 reports on the 19 follow-up studies conducted since 2015 (summarized below) to compare a group of REP subjects who later Transitioned to psychosis (REP-T) with either a group of REP subjects who did Not Transition (REP-NT) or a group of not-at-risk control subjects.

Table 2: Biomarkers significantly related to transition to psychosis resulting from follow-up studies of subjects at risk of psychosis (REP subjects) are presented below with their corresponding effect sizes. Subjects who transitioned to psychosis are labeled REP-T, while those who did not transition to psychosis are labeled REP-NT.

 

REP Subjects

FoU

Psychosis Transition

CT

Group Comparison

(Y)

(n)

REP-T vs. REP-NT

REP-T vs. CT

 

Reference

Biomarker

Def

N

Mean age or Range

 

Yes (n)

No

 

Trend

ES or p- value

Trend

ES or p- value

 

(n)

 

Brain

 

 

 

 

 

 

sMRI

Das et al. (2018) [14]

Brain transi- tivity

ARMS

79

24.4

3.8

16

63

44

Hg= 1.76

Hg= 1.41

 

Sasabayashi et al. (2017) [20]

LGIa

ARMS

90

21.4

4.9

21

69

n/a

p=0.01

n/a

n/a

Koutsouleris et

al. (2015) [21]

Gray matter volumeb

ARMS

73

24.8

4.4

33

40

n/a

PPV=80.6 NPV=81.0

n/a

n/a

Chung et al. (2019) [22]

Brain age gap

CHR

476

12_35

2

67

409

n/a

MuM

HazR=

n/a

n/a

1.21

 

Takayanagi et al. (2017) [23]

Surface areas of the left ACG

 

ARMS

 

73

 

21.7

 

2

 

17

 

56

 

74

n/a

n/a

d=0.69

Thickness of the left ACG

n/a

n/a

d=−0.53

 

rs-fMRI

 

Ilzarbe et al. (2021) [29]

Medical pre- frontal cortex connectivity

 

CHR

 

39

 

15.5

 

1.2

 

15

 

24

 

27

 

p < 0.001 ROC=0.83

Sens=0.79

Sp=0.79

 

 

p = 0.046

 

EEG

Hamilton, Roach et al. (2019a) [27]

Auditory target P3b amplitude

 

PRS

 

298

 

19.2

 

2

 

73

 

225

 

n/a

 

d=0.26 HazR = 1.45

 

n/a

 

n/a

 

Hamilton, Woods et al. (2019b) [28]

Auditory and visual target P3b ampli- tudes

 

PRS

 

33

 

16.2

 

1.8

 

15

 

18

 

n/a

 

 

d = 0.62

HazR = 1.89

 

n/a

 

n/a

 

Bock et al.-2020 [25]

Microstate D coverage

 

UHR

 

54

 

25.5

 

3

 

20

 

34

 

25

d= -0.84

n/a

n/a

Microstate D occurrence

d= -1.24

n/a

n/a

Higuchi et al.

-2021 [24]

P300 latency

 

ARMS

 

33

 

19.2

 

3.5

 

8

 

25

 

28

p= 0.039

n/a

n/a

P300 ampli- tude

ø

ø

p=0.011

Tateno et al. (2021) [26]

dMMN ampli- tude

ARMS

39

18.5

2

11

28

22

p= 0.017

p=0.004

 

 

MEG

 

Grent-‘t-Jong et

al. (2021) [75]

40 Hz ASSR in

right thalamus

 

 

CHR

 

 

116

 

 

22

 

 

1.5

 

 

13

 

 

97

 

 

49

d=0.72

d=1.02

LRM

ROC=0.70

n/a

n/a

40 Hz ASSR in right hippoc- ampus

 

ø

 

ø

 

 

d=0.88

 

fNIRS

Koike et al. (2017) [31]

Centroid value of brain wave- forms

 

UHR

 

34

 

21

 

1

 

6

 

28

 

33

 

n/a

 

n/a

 

MuM

 

Sens= 6/6

 

 

EMG

 

Cadenhead et al. (2020) [32]

 

Acoustic star- tle latency

 

 

CHR

 

 

543

 

 

18.7

 

 

1

 

 

58

 

 

255

 

 

218

 

 

ROC=0.65

 

 

n/a

 

 

n/a

in females

ROC=0.54

in males

 

 

 

Blood

 

Fatty Acids

Clark et al. (2016) [33]

Nervonic acid levels Omega 3d

 

UHR

 

40

 

12.9 - 22.3

 

1

 

11

 

29

 

n/a

 

MuM

Sens =

 

n/a

 

n/a

81.80%

Sp = 78.6%

Inflamma- tory bi- omarker

Föcking et al. (2016) [34]

Interleukin

12/23

 

ARMS

 

39

 

16.1

 

1

 

11

 

28

 

n/a

 

Fold change:

1.57

 

n/a

 

n/a

Ge- netic

 

Jeffries et al. (2016) [35]

Leukocytes miRNA ex- pression

 

CHR

 

67

 

18.4

 

2

 

30

 

37

 

27

 

ROC=

 

ROC=

0.86

0.75

Others

 

ORS

Langbein et al. (2018)

 

NS

 

UHR

 

79

 

20.6

 

1

 

13

 

66

 

180

 

n/a

 

n/a

 

Partial

η2=

 

[36]

0.041

 

Salivary samples

Worthington et

al. (2021) [37]

Salivary cor- tisol

CHR

417

18.7

2

54

363

n/a

HazR = 21.5

n/a

n/a

 

Investigation of brain biomarkers as predictors of transition to psychosis:

In five studies, REP-T and REPNT subjects were compared through structural magnetic resonance imaging (sMRI). The REP-T subjects had: 1- higher brain transitivity (p = 0.042; the same increase was observed in REP-T subjects compared to control subjects; p < 0.001) [14]; 2- higher local gyrification index (LGI) in the left occipital region (p = 0.014) [20]; and 3- decreased gray matter volume (GMV) in prefrontal, perisylvian and subcortical structures (p < 0.001) [21]. Also, in a cohort of 476 REP subjects, a multivariate model involving the brain age gap biomarker was found to be predictive of transition to psychosis (p < 0.05) [22]. Furthermore, although no differences in the anterior cingulate gyrus (ACG) morphology were found between REP-T and REP-NT subjects, compared with control subjects, REP-T subjects showed increased surface areas and decreased thickness of the left ACG (p = 0.025) [23].

In five other studies, REP-T and REP-NT subjects were compared using electroencephalogram (EEG) assessments. The REP-T subjects had: 1- prolonged P300 latencies (p = 0.011 and 0.022) [24], 2- decreased EEG microstate D coverage and occurrence (p = 0.003 and < 0.001, respectively) [25], and 3- reduced amplitude of duration mismatch negativity (dMMN) at baseline (p = 0.017; the same decreased dMMN was observed in REP-T subjects compared to control subjects; p = 0.004) [26]. Additionally, REP-T subjects had a reduced dMMN amplitude at follow-up, compared with the baseline. Furthermore, in a cohort of 298 REP subjects, REP-T subjects were found to have a reduced auditory target P3b amplitude; a smaller auditory target P3b amplitude was also associated with a shorter lead time to the onset of psychosis (p = 0.048) [27]. In a new cohort of 33 REP subjects, Hamilton et al. [28] added the visual modality of target P3b. As a result, they found that both the auditory and visual target P3b amplitudes were reduced in REP-T subjects and predicted lead time to the onset of psychosis (p = 0.002). Along the same lines, greater deficits in target P3b predicted more imminent risks of psychosis onset. Assessment through resting state functional magnetic resonance imaging (rs-fMRI) showed that REP-T subjects had lower medial prefrontal cortex connectivity (p < 0.001 and p = 0.046, respectively) compared with REP-NT subjects and control subjects [29]. At follow-up, it was found that REP subjects with lower connectivity were more likely to transition to psychosis.

Additionally, compared with REP-NT subjects and control subjects, REP-T subjects had impaired and reduced amplitude in the right thalamus during 40 Hz auditory steady- state responses (ASSR) measured with magnetoencephalography (MEG; p = 0.049 and 0.011, respectively) [30]. Moreover, transition to psychosis was predicted by 40 Hz ASSR impairments: ASSR activity in the right thalamus correctly classified 76.9% of the REP-T subjects. Through functional near-infrared spectroscopy (fNIRS) using the timing of frontal activity (Centroid value), all the REP-T subjects (100%; 6/6) were successfully classified into the psychosis spectrum group, whereas 83.3% of them were correctly classified based on the intensity of frontal activity (Integrated value) [31] Finally, compared with electromyography (EMG) assessments of REP-NT subjects, REP-T subjects had longer acoustic startle latencies. This difference in transition groups was more prominent in females (p < 0.05) [32].

Investigation of blood biomarkers as predictors of transition to psychosis: Both omega-3 and nervonic acid levels predicted transition to psychosis (p = 0.023 and 0.041, respectively) [33]. Also, REP-T subjects had increased interleukin 12/23 compared with REP-NT subjects (p = 0.003) [34].

Investigation of genetic biomarkers as predictors of transition to psychosis: Compared with REP-NT subjects and control subjects, REP-T subjects had much weaker miRNA group orchestration (p = 0.012) [35].

Investigation of other types of biomarkers as predictors of transition to psychosis: Although no niacin skin sensitivity (NS) differences were observed between REP-T and REP-NT subjects, REP-T subjects were found to have decreased NS compared with control subjects (p = 0.005, 0.024 and 0.041) [36]. Also, higher levels of salivary cortisol were predictive of transition to psychosis in a cohort of 417 REP subjects, among which 13.2% transitioned to psychosis (p = 0.004) [37].

Biomarkers among REP subjects, before transition to psychosis

As an alternative to relating a biomarker to transition to psychosis, other studies compared REP subjects with not-at-riskcontrol subjects or stratified the group of REP subjects under study into subgroups of subjects with a higher (REP+) and lower level of deficits (REP- ). Table 3 presents a summary of results from 85 studies published since 2015.

Table 3: Below is a distribution of results from studies conducted since January 2015 on biomarkers among subjects at risk of psychosis before transition to psychosis, according to the type of biomarker, study design and group comparison.

 

 

 

 

Biomarkers

 

 

Group comparison

Trend in REP or REP+ subjects in the

biomarker assessment comparison

 

Study

 

Nbr. of stud- ies

 

 

References

 

 

REP vs. CT

 

 

REP+ vs.

REP-

 

 

REP vs.

FEP

 

 

 

 

 

MuM of predic- tion

 

Non-signifi- cant differ- ences

de- sign

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Brain

 

 

 

sMRI

FoU

4

[14,20,38,117]

[20,117]

[38]

[14]

[20,38]

[38]

[38]

[14,117]

 

C-S

 

13

 

[6,39-45,

116,118–121]

 

[6,39-45,

116,118–121]

 

 

[41,43]

 

[41,42]

 

[39,40,43–45]

 

 

[6,43, 116,

118–121]

 

fMRI

FoU

3

[46,47,51]

[46,47,51]

 

 

[46,47]

[46,47,51]

 

 

C-S

4

[48–50,122]

[48–50,122]

 

 

[48]

[48–50]

 

[122]

rs-fMRI

C-S

8

[4,52–58]

[4,53–58]

[52,53]

[58]

[52,54–

57]

[4,53–58]

[55,57,58]

[58]

DTI

C-S

4

[59–61,63]

[59–61,63]

 

 

[59,63]

[59–61]

 

[59]

DWI

C-S

1

[62]

[62]

 

 

 

[62]

 

 

 

 

 

EEG

FoU

7

[25,27, 28, 64,

65,69, 71]

[27,28, 64,65,

69,71]

[64]

[25]

[25,71]

[25,27, 28,64, 65,69,

71]

 

[64,65]

 

C-S

 

11

 

[66–68,70, 72–

74,123–126]

 

[66–68,70,72–

74,123–126]

 

 

 

[72]

 

[66-68,70,72–74]

 

 

[123–126]

 

MEG

C-S

1

[75]

[75]

 

 

 

[75]

 

 

FoU

1

[127]

[127]

 

 

 

 

 

[127]

MRS

C-S

1

[77]

[77]

 

 

[77]

 

 

 

fNIRS

C-S

1

[31]

[31]

 

 

 

 

[31]

 

PET

C-S

2

[76,128]

[128]

[76]

 

 

[76]

 

[128]

EMG

FoU

1

[32]

[32]

 

 

 

 

 

[32]

 

 

 

 

 

 

Blood

Inflamma- tory

C-S

2

[78,79]

[78]

 

[78,79]

[78,79]

[78]

 

[78]

Fatty Acids

C-S

1

[81]

[81]

 

 

[81]

[81]

 

 

Hormonal

C-S

1

[82]

[82]

 

 

[82]

 

 

 

Aminoacids

C-S

1

[66]

[66]

 

 

 

 

 

[66]

Enzymes

FoU

2

[80,83]

[80,83]

 

[80]

[80]

[83]

 

[80]

Protein

FoU

1

[129]

[129]

 

 

 

 

 

[129]

 

 

Ge-

netic

NRG1

mRNA

FoU

1

[85]

 

[85]

 

[85]

 

 

 

TSPO

rs6971

C-S

1

[130]

[130]

 

 

 

 

[130]

 

PSRS

C-S

1

[86]

 

 

[86]

[86]

 

 

 

 

 

 

 

 

 

Eyes

 

Oculomotor abnormali- ties

 

 

C-S

 

 

4

 

[87–

89,131,132]

 

[87–

89,131,132]

 

 

[87,88]

 

 

 

[87–89]

 

 

 

[87, 88, 131,

132]

 

Retinal ab- normality

 

C-S

 

1

 

[90]

 

 

[90]

 

[90]

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Others

 

Sleep (Ac- tigraphy or sleep high density- EEG)

 

 

 

FoU

 

 

 

1

 

 

 

[91]

 

 

 

[91]

 

 

 

 

[91]

 

 

 

 

C-S

1

[92]

[92]

 

[92]

 

 

 

 

ORS

FoU

1

[36]

[36]

 

 

 

[36]

 

[36]

Fecal sam- ples

C-S

1

[77]

[77]

[77]

[77]

 

 

 

 

Urinary samples

C-S

1

[95]

[95]

 

[95]

 

[95]

 

 

 

ECG or pho- toplethys- mography

 

 

C-S

 

 

2

 

 

[93,94]

 

 

[93,94]

 

 

 

[93]

 

 

 

[93]

 

 

 

[94]

Immu- noassays

C-S

1

[133]

[133]

 

 

 

 

 

[133]

Brain biomarkers:

Structural magnetic resonance imaging (sMRI) assessments: Kambeitz-Ilankovic et al. [38] stratified REP subjects according to their long-term functioning levels: REPs+ (low-functioning level REP subjects, i.e. GAF score < 70) versus REPs- (high-functioning level REP subjects, i.e. GAF score ≥ 70). The REP+ group had: 1- increased surface areas of the left precentral (p < 0.001), lateral occipital cortices (p = 0.032), right superior temporal (p < 0.001) and lateral occipital cortices on the right hemisphere (p < 0.001); 2- reduced cortical areas on the left hemisphere (p between 0.000 and 0.035); and 3- reduced surface values on the right hemisphere (p between 0.000 and 0.032). Furthermore, at follow-up, the neuroanatomical predictor distinguished REP+ from REP- (PPV = 78.6; NPV = 84.6).

Compared with control subjects, REP subjects showed: 1- increased local gyrification index (LGI) in bilateral frontal (p = 0.0001), temporal (p = 0.0448), parietal (p = 0.0001) and occipital (p = 0.0001) regions [20] (LGI was also higher in REP-T subjects compared with REP-NT subjects; Table 2); 2- decreased cortical gyrification, including (a) the LGI in the lateral orbitofrontal superior bank of the superior temporal sulcus, anterior isthmus of the cingulate and temporal poles (p < 0.05); (b) the mean curvature index in the cingulate, post-central and lingual gyrus (p < 0.05); and (c) the sulcal depth in parietal, superior temporal sulcus and cingulate regions (p < 0.05) [39]; 3- lower cortical gyrification (hypogyria) in clusters, including medial parietooccipital and cingulate regions (p < 0.001) [40]; 4- larger white matter volumes in the left Crus I/II, but only in males (p = 0.031; g = 0.856; male REP subjects had also higher white matter volumes in the right Crus I/II compared with subjects with first-episode psychosis (FEP; p = 0.007; g= 1.248) [41]; 5- larger putamen (p < 0.001) [42]; 6- decreased volume of the amygdala lateral nucleus (p = 0.0006) [43]; 7- decreased pineal gland volume (p = 0.015; d = -0.38 to -0.54) [44]; and 8- decreased cortical thickness in the left prefrontal cortex (PFC; p < 0.001), the right PFC (p < 0.001), the left inferior parietal lobule (IPL; p = 0.018) and the right IPL (p = 0.043) [45].

Functional magnetic resonance imaging (fMRI) assessments: Compared with control subjects, REP subjects had: 1- differences over time in connectivity between Crus I and the occipital cortex (p = 0.008) [46]; 2- increased hippocampus/ amygdala activity during neutral faces processing (d = 0.987) [47]; 3- reduced dorsolateral prefrontal activity during failed inhibition (p < 0.05; d = 0.980) [47]; 4- greater probabilistic category learning task activity (i.e. the Weather Prediction Task) in several parietal, occipital and one temporal regions (p < 0.01) [48]; 5- decreased striatal activation (p < 0.01; d = 0.86), activation in the associative striatum (p < 0.01; d = 0.89) and activity in multiple cortical regions connected to the associative striatum (p < 0.01; d= 0.70 to 0.89) [48]; 6- lower brain activation in face processing areas (p = 0.003) [49]; 7- lower activity of a network involving the bilateral auditory cortices, the thalamus and frontal brain regions, mediated by gamma oscillation (p < .01) [50]; and 8- lower brain activation in the left (p = 0.04; d = 0.58) and right caudate (p = 0.04; d = 0.57) [51], decreased activation in the associative cortico-striatal network being negatively associated with variability in the grip force in REP subjects.

Resting state functional magnetic resonance imaging (rsfMRI) assessments: Decross et al. [52] stratified REP subjects according to their level of delusional beliefs: 43 REPs+ (high level) versus 44 REPs- (low level). REPs+ had greater connectivity between the amygdala and visual cortex (p < 0.001). Likewise, Wang et al. [53] stratified REP subjects according to their level of subthreshold psychotic experiences: 22 REPs+ (moderately elevated level) versus 22 REPs- (consistently low level). REPs+ had lower rates of moment-to-moment engagement of brain networks involving the visual and salience networks (p < 0.001; the same result was observed compared with control subjects)

Compared with control subjects, REP subjects showed: 1- increased medial prefrontal cortex (mPFC) – posterior cingulate cortex (PCC) connectivity in resting-state analyses (p = 0.0037), but decreased mPFC–PCC connectivity in task connectivity analyses (p = 0.03) [54]; 2- increased functional connectivity strength (FCS) in the left calcarine cortex, but decreased FCS in the left middle frontal gyrus (accuracy, sensitivity and specificity, respectively: 87.3%, 73.5% and 100%) [55]; 3- increased global efficiency and resilience to targeted attack (p < 0.05), but decreased clustering coefficient (brain functional networks; p < 0.05) [56]; 4- increased regional homogeneity (ReHo) in the right inferior frontal gyrus and the right putamen (p < 0.005; accuracy, sensitivity and specificity, respectively: 90.1%, 88.2% and 91.9%), but decreased ReHo in the left inferior temporal gyrus (p < 0.005) [57]; 5- decreased functional connectivity with nine clusters located at bilateral inferior temporal gyrus (ITG), bilateral transverse temporal gyrus of Heschl, left parahippocampal gyrus, right hippocampus, right thalamus, bilateral cerebellar Crus I/II and right posterior ITG (p < 0.05) [4]; and 6- decreased parameter of asymmetry in the left thalamus (p = 0.001; accuracy, sensitivity and specificity, respectively: 62.1%, 81.1% and 42.3%) [58].

Diffusion tensor imaging (DTI) and Diffusion-weighted imaging (DWI) assessments: Compared with control subjects, REP subjects had: 1- higher radial diffusivity (p = 0.030) and trace (p = 0.031) in the cingulum bundle, but a lower fractional anisotropy (p = 0.028) [59]; 2- lower scores of the area under the rich-club curve, the mean strength of rich-club connections and local efficiency of the right accumbens (p = 0.012) [60]; 3- decreased forceps minor fractional anisotropy and superior longitudinal fasciculus radial diffusivity (p < 0.001) [61]; 4- decreased thalamo-orbitofrontal connectivity (p < 0.05) [62]; and 5- hemispheric asymmetric deficits of nodal efficiency, global and local efficiency (p < 0.05) [63].

Electroencephalogram (EEG) and Magnetoencephalography (MEG) assessments: Fujioka et al. [64] stratified REP subjects according to their remission: REPs+ (non-remitters) versus REPs- (remitters, defined by a GAF score ≥ 61 and a score ≤ 2 on all SOPS positive subscales). At baseline, REP+ subjects showed smaller duration mismatch negativity (dMMN) amplitudes (p = 0.039).

With regard to control subjects, the REP subjects showed: 1- reduced dMMN amplitudes (p = 0.003 [64]; p = 0.02 [65]; p = 0.02 and d = 0.88 [66]; p between 0.005 and 0.018 [67]) and a difference in waveforms MMN (p < 0.05) [68]; 2- reduced 40 Hz auditory steady-state response (ASSR; p < 0.05 [68]; p = 0.04 and d = -0.72 [69]) and late-latency ASSRs (p = 0.02) [70]; 3- increased P50 ratio (p = 0.03), but decreased C-T difference (p= 0.009) [71]; 4- increased P300 inter-trial variability (p = 0.028) [72] and decreased P300 peak amplitudes (p = 0.024 [73]; p = 0.001 [72]); 5- reduced target P3b and novelty P3a amplitudes (p < 0.001; d = 0.37 [27]; p between 0.0002 and 0.006; d = 0.71 to 1.16 [28]); 6- reduced N100 adaptation (p = 0.001) [74]; and 7- decreased phase consistency of β/γ- band oscillations in visual cortex (p = 0.005; d = 0.63) [75].

Magnetic resonance spectroscopy (MRS), functional near-infrared spectroscopy (fNIRS), and Position emission tomography (PET) assessments: Schifani et al. [76] stratified REP subjects according to their levels of stress-induced dopamine release in PFC: REPs+ (higher level) versus REPs- (lower level). REPs+ had lower translocator protein (TSPO) expression in the hippocampus (p = 0.03).Among 47 REP subjects, 82.4% and 70.2% were successfully classified using a modified integral or a centroid value from the frontal area, respectively [31]. Compared with control subjects, REP subjects were found to have increased levels of choline in anterior cingulate (p = 0.03) [77]

Blood biomarkers: Compared with another cohort of first-episode psychosis (FEP) subjects, REP subjects had: 1- lower homocysteine levels and higher methionine/homocysteine ratio (p = 0.016, whereas no differences were observed between REP and control subjects) [78]; 2- higher levels of immunoinflammatory analytes MCP-1, MIP-1β, TARC, BDNF, Eotaxin-1, and IFN- γ (p < 0.001; same results were observed compared with control subjects) [79]; and 3- higher Ndel1 enzymes (p = 0.023; 52= 0.127, whereas no differences were observed between REP and control subjects) [80]

With regard to control subjects, REP subjects had: 1- lower levels of phosphatidylethanolamine and polyunsaturated fatty acids (eicosapentaenoic acid, docosahexaenoic acid, arachidonic acid and Omega-3 index) and higher concentrations of sphingomyelin and nervonic acid (p < 0.0001) [81]; 2- increased levels of serum leptin (p= 0.025) [82]; and 3- lower circulating concentrations of arachidonylethanolamide (p = 0.003) and 2-arachidonoylglycerol (p < 0.001) [83]

Genetic biomarkers: Jagannath et al. [85] stratified REP subjects according to their long-term functioning levels: 98 REPs+ (low level, i.e. GAF score ≤ 64) versus 31 REPs- (high level, i.e. GAF score ≥ 65). After a one-year follow-up, REPs+ had higher pan-NRG1 mRNA (sensitivity = 73.3%; specificity = 57.7%). In REP subjects, the polygenic schizophrenia-related risk score was: 1- negatively associated with hippocampal volumes (p = 0.01) [86]; and 2- positively correlated with the hemispheric asymmetric deficits of local efficiency (p < 0.05) [63].

Eye biomarkers: Caldani et al. [87] stratified REP subjects according to their levels of neurological soft signs: REPs+ (high level) versus REPs- (low level). Compared with control subjects, REPs+ had more errors in the memory-guided saccades (p < 0.002) and more intrusive saccades (p < 0.005) [88]. Also in comparison with control subjects, REP subjects had higher antisaccade error rates (p < 0.001) [89].

Peredo et al. [90] found two clusters according to the REP subjects’ retinal responses to luminance with the electroretinography (ERG), including: a first subgroup with alterations (54 REP+ ) and a second with a control-like ERG profile (53 REP- ). Compared with REPs- , REPs+ were 2.7 more likely to present impaired cognitive function (p = 0.001).

Other types of biomarkers: He et al. [77] stratified REP subjects according to the definition of their risk of psychosis: REPs+ (UHRs) versus REPs- (HR). REPs+ had increased levels of orders Clostridiales, Lactobacillales,, and Bacteroidales in fecal samples (p = 0.048; same results were observed compared with control subjects).

Compared with control subjects, REP subjects showed: 1) increased fragmented circadian rhythms and later onset of nocturnal rest (p = 0.04) [91]; 2- more wakefulness after sleep onset (p = 0.048; d = 0.63) and higher non-rapid eye movement sleep EEG power in the gamma band (p = 0.012; d = 0.816), observed in a large fronto-parieto-occipital area [92]; 3- decreased niacin skin sensitivity (NS; p < 0.05; when REP subjects were defined with attenuated symptoms, whereas no group differences were found between REP subjects defined with genetic risk and control subjects) [36]; 4- reduced variance of resting heart rate variability (HRV; p = 0.004; d = -0.74) and a reduced high-frequency power (p = 0.024; d = -0.64) during deep breathing [93]; however, Clamor et al. [94] found no differences in HRV; and 5- increase in biopyrrins and reduction of free immunoglobin light chains K and λ in urinary samples (p = 0.035) [95].

REFERENCES

1. Fujino H, Sumiyoshi C, Yasuda Y, Yamamori H, Fujimoto M, Fukunaga M, et al. Estimated cognitive decline in patients with schizophrenia: A multicenter study. Psychiatry Clin Neurosci. 2017; 71: 294-300.

2. American Psychiatric Association. Diagnostic and statistical manual of mental disorders: DSM-5. 2013.

3. Perkins DO, Gu H, Boteva K, Lieberman JA. Relationship between duration of untreated psychosis and outcome in first-episode schizophrenia: A critical review and meta-analysis. Am J Psychiatry. 2005; 162: 1785–1804.

4. Anteraper SA, Collin G, Guell X, Scheinert T, Molokotos E, Henriksen MT, et al. Altered resting-state functional connectivity in young children at familial high risk for psychotic illness: A preliminary study. Schizophr Res. 2020; 216: 496-503.

5. Fusar-Poli P, Borgwardt S, Crescini A, Deste G, Kempton MJ, Lawrie S, et al. Neuroanatomy of vulnerability to psychosis: A voxel-based metaanalysis. Neurosci Biobehav Rev [Internet]. 2011; 35: 1175-1185.

6. Hwang WJ, Cho KIK, Kwak YB, Lee J, Kim M, Lee TY, et al. Intact thalamic microstructure in asymptomatic relatives of schizophrenia patients with high genetic loading. Schizophr Res. 2020; 230: 111-113.

7. Hans SL, Auerbach JG, Styr B, Marcus J. Offspring of parents with schizophrenia: Mental disorders during childhood and adolescence. Schizophr Bull. 2004; 30: 303-315.

8. Niemi LT, Suvisaari JM, Tuulio-Henriksson A, Lonnqvist JK. Childhood developmental abnormalities in schizophrenia: evidence from highrisk studies. Schizophr Res. 2003; 60: 239-258.

9. Sandstrom A, Sahiti Q, Pavlova B, Uher R. Offspring of parents with schizophrenia, bipolar disorder, and depression: A review of familial high-risk and molecular genetics studies. Psychiatr Genet. 2020: 160- 169.

10.Liu CH, Keshavan MS, Tronick E, Seidman LJ. Perinatal Risks and Childhood Premorbid Indicators of Later Psychosis: Next Steps for Early Psychosocial Interventions. Schizophr Bull. 2015; 41: 801-816.

11.Yung AR, Yuen HP, McGorry PD, Phillips LJ, Kelly D, Dell’Olio M, et al. Mapping the onset of psychosis: the Comprehensive Assessment of At-Risk Mental States. Aust N Z J Psychiatry [Internet]. 2005; 39: 964-971.

12.Rasic D, Hajek T, Alda M, Uher R. Risk of mental illness in offspring of parents with schizophrenia, bipolar disorder, and major depressive disorder: A meta- analysis of family high-risk studies. Schizophr Bull. 2014; 40: 28-38.

13.Fusar-Poli P, Borgwardt S, Bechdolf A, Addington J, RiecherRössler A, Schultze- Lutter F, et al. The psychosis high-risk state: A comprehensive state-of-the-art review. Arch Gen Psychiatry. 2013; 70: 107-120.

14.Das T, Borgwardt S, Hauke DJ, Harrisberger F, Lang UE, RiecherRössler A, et al. Disorganized gyrification network properties during the transition to psychosis. JAMA Psychiatry. 2018; 75: 613-622.

15.Stafford MR, Jackson H, Mayo-Wilson E, Morrison AP, Kendall T. Early interventions to prevent psychosis: systematic review and metaanalysis. BMJ. 2013; 346: f185.

16.Atkinson AJ, Colburn WA, DeGruttola VG, DeMets DL, Downing GJ, Hoth DF, et al. Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001; 69: 89-95.

17.Pickard BS. Schizophrenia biomarkers: Translating the descriptive into the diagnostic. J Psychopharmacol. 2015; 29: 138-143.

18.Weickert CS, Weickert TW, Pillai A, Buckley PF. Biomarkers in schizophrenia: a brief conceptual consideration. Dis Markers. 2013; 35: 3-9.

19.Munn Z, Peters MDJ, Stern C, Tufanaru C, McArthur A, Aromataris E. Systematic review or scoping review? Guidance for authors when choosing between a systematic or scoping review approach. BMC Med Res Methodol. 2018; 18: 1-7.

20.Sasabayashi D, Takayanagi Y, Takahashi T, Koike S, Yamasue H, Katagiri N, et al. Increased Occipital Gyrification and Development of Psychotic Disorders in Individuals With an At-Risk Mental State: A Multicenter Study. Biol Psychiatry. 2017; 82: 737-745.

21.Koutsouleris N, Riecher-Rössler A, Meisenzahl EM, Smieskova R, Studerus E, Kambeitz-Ilankovic L, et al. Detecting the Psychosis Prodrome Across High-Risk Populations Using Neuroanatomical Biomarkers. Schizophr Bull. 2015; 41: 471-4 82.

22.Chung Y, Addington J, Bearden CE, Cadenhead K, Cornblatt B, Mathalon DH, et al. Adding a neuroanatomical biomarker to an individualized risk calculator for psychosis: A proof-of-concept study. Schizophr Res. 2019; 208: 41-43.

23.Takayanagi Y, Kulason S, Sasabayashi D, Takahashi T, Katagiri N, Sakuma A, et al. Reduced Thickness of the Anterior Cingulate Cortex in Individuals with an At- Risk Mental State Who Later Develop Psychosis. Schizophr Bull. 2017; 43: 907-9 13.

24.Higuchi Y, Sumiyoshi T, Tateno T, Nakajima S, Sasabayashi D, Nishiyama S, et al. Prolonged p300 latency in antipsychotic-free subjects with at-risk mental states who later developed schizophrenia. J Pers Med. 2021; 11.

25.Bock R De, Mackintosh AJ, Maier F, Borgwardt S, Riecher-rössler A, Andreou C. EEG microstates as biomarker for psychosis in ultra-highrisk patients. Transl Psychiatry. 2020; 10: 1-9.

26.Tateno T, Higuchi Y, Nakajima S, Sasabayashi D, Nakamura M, Ueno M, et al. Features of Duration Mismatch Negativity around the Onset of Overt Psychotic Disorders: A Longitudinal Study. Cereb Cortex. 2021; 31: 2416-2424.

27.Hamilton HK, Roach BJ, Bachman PM, Belger A, Carrion RE, Duncan E, et al. Association between P300 Responses to Auditory Oddball Stimuli and Clinical Outcomes in the Psychosis Risk Syndrome. JAMA Psychiatry. 2019; 76: 1187-11 97.

28.Hamilton HK, Woods SW, Roach BJ, Llerena K, McGlashan TH, Srihari VH, et al. Auditory and Visual Oddball Stimulus Processing Deficits in Schizophrenia and the Psychosis Risk Syndrome: Forecasting Psychosis Risk with P300. Schizophr Bull. 2019; 45: 1068-1080.

29.Ilzarbe D, Baeza I, de la Serna E, Fortea A, Valli I, Puig O, et al. Theory of mind performance and prefrontal connectivity in adolescents at clinical high risk for psychosis. Dev Cogn Neurosci. 2021; 48: 100940.

30.Grent-‘t-Jong T, Gajwani R, Gross J, Gumley AI, Krishnadas R, Lawrie SM, et al. 40-Hz Auditory Steady-State Responses Characterize Circuit Dysfunctions and Predict Clinical Outcomes in Clinical High-Risk for Psychosis Participants: A Magnetoencephalography Study. Biol Psychiatry. 2021; 1-11.

31.Koike S, Satomura Y, Kawasaki S, Nishimura Y, Kinoshita A, Sakurada H, et al. Application of functional near infrared spectroscopy as supplementary examination for diagnosis of clinical stages of psychosis spectrum. Psychiatry Clin Neurosci. 2017; 71: 794-806.

32.Cadenhead KS, Duncan E, Addington J, Bearden C, Cannon TD, Cornblatt BA, et al. Evidence of Slow Neural Processing, Developmental Differences and Sensitivity to Cannabis Effects in a Sample at Clinical High Risk for Psychosis From the NAPLS Consortium Assessed With the Human Startle Paradigm. Front Psychiatry. 2020; 11: 1–11.

33.Clark SR, Baune BT, Schubert KO, Lavoie S, Smesny S, Rice SM, et al. Prediction of transition from ultra-high risk to first-episode psychosis using a probabilistic model combining history, clinical assessment and fatty-acid biomarkers. Transl Psychiatry. 2016; 6: e897.

34.Föcking M, Dicker P, Lopez LM, Cannon M, Schäfer MR, McGorry PD, et al. Differential expression of the inflammation marker IL12p40 in the at-risk mental state for psychosis: A predictor of transition to psychotic disorder? BMC Psychiatry. 2016; 16: 1-8.

35.Jeffries CD, Perkins DO, Chandler SD, Stark T, Yeo E, Addington J, et al. Insights into psychosis risk from leukocyte microRNA expression. Transl Psychiatry. 2016; 6.

36.Langbein K, Schmidt U, Schack S, Biesel NJ, Rudzok M, Amminger GP, et al. State marker properties of niacin skin sensitivity in ultra-high risk groups for psychosis - An optical reflection spectroscopy study. Schizophr Res. 2018; 192: 377-384.

37.Worthington MA, Walker EF, Addington J, Bearden CE, Cadenhead KS, Cornblatt BA, et al. Incorporating cortisol into the NAPLS2 individualized risk calculator for prediction of psychosis. Schizophr Res. 2021; 227: 95-100.

38.Kambeitz-Ilankovic L, Meisenzahl EM, Cabral C, von Saldern S, Kambeitz J, Falkai P, et al. Prediction of outcome in the psychosis prodrome using neuroanatomical pattern classification. Schizophr Res. 2016; 173: 159-165.

39.Damme KS, Gupta T, Nusslock R, Bernard JA, Orr JM, Mittal VA. Cortical morphometry in the psychosis risk period: A comprehensive perspective of surface features. Biol Psychiatry Cogn Neurosci Neuroimaging. 2019; 4: 434-443.

40.Park I, Kim M, Lee TY, Hwang WJ, Bin Kwak Y, Oh S, et al. Reduced cortical gyrification in the posteromedial cortex in unaffected relatives of schizophrenia patients with high genetic loading. npj Schizophr. 2021; 7: 1-8.

41.Morimoto C, Uematsu A, Nakatani H, Takano Y. Volumetric differences in gray and white matter of cerebellar Crus I/II across the different clinical stages of schizophrenia. Psychiatry Clin Neurosci. 2021; 75: 256-264.

42.Gong Q, Scarpazza C, Dai J, He M, Xu X, Shi Y, et al. A transdiagnostic neuroanatomical signature of psychiatric illness. Neuropsychopharmacology. 2019; 44: 869-875.

43.Armio RL, Laurikainen H, Ilonen T, Walta M, Salokangas RKR, Koutsouleris N, et al. Amygdala subnucleus volumes in psychosis high-risk state and first-episode psychosis: Amygdala subnuclei and psychosis. Schizophr Res. 2020; 215: 284-292.

44.Takahashi T, Nakamura M, Sasabayashi D, Nishikawa Y, Takayanagi Y, Nishiyama S, et al. Reduced pineal gland volume across the stages of schizophrenia. Schizophr Res. 2019; 206: 163-170.

45.Kwak YB, Kim M, Cho KIK, Lee J, Lee TY, Kwon JS. Reduced cortical thickness in subjects at clinical high risk for psychosis and clinical attributes. Aust New Zeal J Psychiatry. 2019; 53: 219-227.

46.Bernard JA, Orr JM, Mittal VA. Cerebello-thalamo-cortical networks predict positive symptom progression in individuals at ultra-high risk for psychosis. NeuroImage Clin. 2017; 14: 622-628.

47. Bourque J, Spechler PA, Potvin S, Whelan R, Banaschewski T, Bokde AL, et al. Functional neuroimaging predictors of self-reported psychotic symptoms in adolescents. Am J Psychiatry. 2017; 174: 566-575.

48. Kraker NR, Hua JPY, and Kerns JG. Probabilistic Category Learning and Striatal Functional Activation in Psychosis Risk. Schizophr Bull. 2019; 45: 396-404.

49. Spilka MJ, Arnold AE, Goghari VM. Functional activation abnormalities during facial emotion perception in schizophrenia patients and nonpsychotic relatives. Schizophr Res. 2015; 168: 330-337.

50. Leicht G, Vauth S, Polomac N, Andreou C, Rauh J, Mußmann M, et al. EEG-Informed fMRI Reveals a Disturbed Gamma-Band-Specific Network in Subjects at High Risk for Psychosis. Schizophr Bull. 2016; 42: 239–249.

51. Dean DJ, Bernard JA, Damme KSF, Reilly RO, Orr JM, Mittal VA. Longitudinal Assessment and Functional Neuroimaging of Movement Variability Reveal Novel Insights Into Motor Dysfunction in Clinical High Risk for Psychosis. 2020; 46: 1567–1576.

52. Decross SN, Farabaugh AH, Holmes AJ, Ward M, Boeke EA, Wolthusen RPF, et al. Increased amygdala-visual cortex connectivity in youth with persecutory ideation. Psychol Med. 2019; 50: 273-283.

53. Wang D, Peng X, Pelletier-Baldelli A, Orlov N, Farabaugh A, Nasr S, et al. Altered temporal, but intact spatial, features of transient network dynamics in psychosis. Mol Psychiatry [Internet]. 2021; 26: 2493–2503.

54. Damme KSF, Pelletier-Baldelli A, Cowan HR, Orr JM, Mittal VA. Distinct and opposite profiles of connectivity during self-reference task and rest in youth at clinical high risk for psychosis. Hum Brain Mapp. 2019; 40: 3254–3264.

55.Li RR, Lyu HL, Liu F, Lian N, Wu RR, Zhao JP, et al. Altered functional connectivity strength and its correlations with cognitive function in subjects with ultra-high risk for psychosis at rest. CNS Neurosci Ther. 2018; 24: 1140-1148.

56. Lo CYZ, Su TW, Huang CC, Hung CC, Chen WL, Lan TH, et al. Randomization and resilience of brain functional networks as systems-level endophenotypes of schizophrenia. Proc Natl Acad Sci U S A. 2015; 112: 9123–9128.

57.Wang S, Wang G, Lv H, Wu R, Zhao J, Guo W. Abnormal regional homogeneity as potential imaging biomarker for psychosis risk syndrome: A resting-state fMRI study and support vector machine analysis. Sci Rep. 2016; 6: 1-8.

58. Zhu F, Liu Y, Liu F, Yang R, Li H, Chen J, et al. Functional asymmetry of thalamocortical networks in subjects at ultra-high risk for psychosis and first-episode schizophrenia. Eur Neuropsychopharmacol. 2019; 29: 519–528.

59. Fitzsimmons J, Rosa P, Sydnor VJ, Reid BE, Makris N, Goldstein JM, et al. Cingulum bundle abnormalities and risk for schizophrenia. Schizophr Res. 2020; 215: 385-391.

60. Schmidt A, Crossley NA, Harrisberger F, Smieskova R, Lenz C, Riecher-Rössler A, et al. Structural network disorganization in subjects at clinical high risk for psychosis. Schizophr Bull. 2017; 43: 583-591.

61. Prasad KM, Upton CH, Schirda CS, Nimgaonkar VL., Keshavan MS. White matter diffusivity and microarchitecture among schizophrenia subjects and first-degree relatives. Schizophr Res. 2015; 161: 70-75.

62. Cho KIK, Shenton ME, Kubicki M, Jung WH, Lee TY, Yun JY, et al. Altered thalamo-cortical white matter connectivity: probability tractography study in clinical-high risk for psychosis and first-episode psychosis. Schizophr Bull. 2016; 42: 723-731. 

63.Zhu Y, Wang S, Gong X, Edmiston EK, Zhong S, Li C, et al. Associations between hemispheric asymmetry and schizophrenia-related risk genes in people with schizophrenia and people at a genetic high risk of schizophrenia. Br J Psychiatry. 2021; 219: 392-400.

64.Fujioka M, Kirihara K, Koshiyama D, Tada M, Nagai T, Usui K, et al. Mismatch Negativity Predicts Remission and Neurocognitive Function in Individuals at Ultra- High Risk for Psychosis. Front Psychiatry. 2020; 11: 1-10.

65.Koshiyama D, Kirihara K, Tada M, Nagai T, Koike S, Suga M, et al. Duration and frequency mismatch negativity shows no progressive reduction in early stages of psychosis. Schizophr Res. 2017; 190: 32- 38.

66.Nagai T, Kirihara K, Tada M, Koshiyama D, Koike S, Suga M, et al. Reduced Mismatch Negativity is Associated with Increased Plasma Level of Glutamate in First-episode Psychosis. Sci Rep. 2017; 7: 1-9.

67.Pantlin LN, Davalos D. Neurophysiology for Detection of High Risk for Psychosis. Schizophr Res Treatment. 2016.

68.Adams R, Pinotsis D, Tsirlis K, Ji JL, Repovs G, Murray J, et al. Computational Modelling of EEG and fMRI Paradigms Reveals a Consistent Loss of Pyramidal Cell Synaptic Gain in Schizophrenia. Biol Psychiatry. 2021; 89: S95.

69.Koshiyama D, Kirihara K, Tada M, Nagai T, Fujioka M, Ichikawa E, et al. Auditory gamma oscillations predict global symptomatic outcome in the early stages of psychosis: A longitudinal investigation. Clin Neurophysiol. 2018; 129: 2268-2275.

70.Tada M, Nagai T, Kirihara K, Koike S, Suga M, Araki T, et al. Differential Alterations of Auditory Gamma Oscillatory Responses between PreOnset High- Risk Individuals and First-Episode Schizophrenia. Cereb Cortex. 2016; 26: 1027-10 35.

71.Shaikh M, Dutt A, Broome MR, Vozmediano AG, Ranlund S, Diez A, et al. Sensory gating deficits in the attenuated psychosis syndrome. Schizophr Res. 2015; 161: 277-282.

72.Kim M, Lee TH, Kim JH, Hong H, Lee TY, Lee Y, et al. Decomposing P300 into correlates of genetic risk and current symptoms in schizophrenia: An inter-trial variability analysis. Schizophr Res. 2018; 192: 232-239.

73.Graber K, Bosquet Enlow M, Duffy FH, D’Angelo E, Sideridis G, Hyde DE, et al. P300 amplitude attenuation in high risk and early onset psychosis youth. Schizophr Res. 2019; 210: 228-238.

74.Gonzalez-Heydrich J, Bosquet Enlow M, D’Angelo E, Seidman LJ, Gumlak S, Kim A, et al. N100 Repetition Suppression Indexes Neuroplastic Defects in Clinical High Risk and Psychotic Youth. Neural Plast. 2016; 2016.

75.Grent-’t-Jong T, Gajwani R, Gross J, Gumley AI, Krishnadas R, Lawrie SM, et al. Association of Magnetoencephalographically Measured HighFrequency Oscillations in Visual Cortex with Circuit Dysfunctions in Local and Large-scale Networks during Emerging Psychosis. JAMA Psychiatry. 2020; 1-11.

76.Schifani C, Hafizi S, Tseng HH, Gerritsen C, Kenk M, Wilson AA, et al. Preliminary data indicating a connection between stress-induced prefrontal dopamine release and hippocampal TSPO expression in the psychosis spectrum. Schizophr Res. 2018; 213: 80-86.

77.He Y, Kosciolek T, Tang J, Zhou Y, Li Z, Ma X, et al. Gut microbiome and magnetic resonance spectroscopy study of subjects at ultra-high risk for psychosis may support the membrane hypothesis. Eur Psychiatry. 2018; 53: 37-45

78.Onozato M, Uta A, Magarida A, Fukuoka N, Ichiba H, Tsujino N, et al. Alterations in methionine to homocysteine ratio in individuals with first-episode psychosis and those with at-risk mental state. Clin Biochem. 2020; 77(September 2019): 48-53.

79.Kelsven S, de la Fuente-Sandoval C, Achim CL, Reyes-Madrigal F, Mirzakhanian H, Domingues I, et al. Immuno-inflammatory changes across phases of early psychosis: The impact of antipsychotic medication and stage of illness. Schizophr Res. 2020; 226: 13-23.

80.Dal Mas C, Nani J V., Noto C, Yonamine CM, da Cunha GR, Mansur RB, et al. Ndel1 oligopeptidase activity as a potential biomarker of early stages of schizophrenia. Schizophr Res. 2019; 208: 202-208.

81.Alqarni A, Mitchell TW, McGorry PD, Nelson B, Markulev C, Yuen HP, et al. Comparison of erythrocyte omega-3 index, fatty acids and molecular phospholipid species in people at ultra-high risk of developing psychosis and healthy people. Schizophr Res. 2019; 226: 44-51.

82.Martorell L, Muntané G, Porta-López S, Moreno I, Ortega L, Montalvo I, et al. Increased levels of serum leptin in the early stages of psychosis. J Psychiatr Res. 2019; 111: 24-29.

83.Joaquim HPG, Costa AC, Pereira CAC, Talib LL, Bilt MMV, Loch AA, et al. Plasmatic endocannabinoids are decreased in subjects with ultra-high risk of psychosis. Eur J Neurosci. 2022; 55: 1079-1087.

84.Jagannath V, Gerstenberg M, Walitza S, Franscini M, Heekeren K, Rössler W, et al. Neuregulin 1 (NRG1) gene expression predicts functional outcomes in individuals at clinical high-risk for psychosis. Psychiatry Res. 2018; 266: 143-146.

85.Harrisberger F, Smieskova R, Vogler C, Egli T, Schmidt A, Lenz C, et al. Impact of polygenic schizophrenia-related risk and hippocampal volumes on the onset of psychosis. Transl Psychiatry. 2016; 6: e868- 874.

86.Caldani S, Bucci MP, Lamy JC, Seassau M, Bendjemaa N, Gadel R, et al. Saccadic eye movements as markers of schizophrenia spectrum: Exploration in at-risk mental states. Schizophr Res. 2016; 181: 30-37.

87.Caldani S, Amado I, Bendjemaa N, Vialatte F, Mam-Lam-Fook C, Gaillard R, et al. Oculomotricity and Neurological Soft Signs: Can we refine the endophenotype? A study in subjects belonging to the spectrum of schizophrenia. Psychiatry Res. 2017; 256: 490-497.

88.Türközer HB, Ivleva EI, Palka J, Clementz BA, Shafee R, Pearlson GD, et al. Biomarker Profiles in Psychosis Risk Groups Within Unaffected Relatives Based on Familiality and Age. Schizophr Bull. 2021; 1-10.

89.Peredo R, Gagné AM, Gilbert E, Hébert M, Maziade M, Mérette C. Electroretinography may reveal cognitive impairment among a cohort of subjects at risk of a major psychiatric disorder. Psychiatry Res. 2020; 291: 113227.

90.Lunsford-Avery JR, Gonçalves BDSB, Brietzke R, Bressan RA, Gadelha A, Auerbach RP, et al. Adolescents at Clinical-High Risk for Psychosis: Circadian Rhythm Disturbances Predict Worsened Prognosis at 1-Year Follow-up. Schizophr Res. 2017; 189: 37-42.

91.Mayeli A, LaGoy A, Donati FL, Kaskie RE, Najibi SM, Ferrarelli F. Sleep abnormalities in individuals at clinical high risk for psychosis. J Psychiatr Res. 2021; 137: 328-334.

92.Liu YW, Tzeng NS, Yeh C Bin, Kuo TBJ, Huang SY, Chang CC, et al. Reduced cardiac autonomic response to deep breathing: A heritable vulnerability trait in patients with schizophrenia and their healthy first-degree relatives. Psychiatry Res. 2016; 243: 335-341.

93.Clamor A, Ludwig L, Lincoln TM. Heart rate variability as an index of emotion (dys) regulation in psychosis? 2020; 158: 310-317.

94.Wake R, Araki T, Fukushima M, Matsuda H, Inagaki T, Hayashida M, et al. Urinary biopyrrins and free immunoglobin light chains are biomarker candidates for screening at-risk mental state in adolescents. Early Interv Psychiatry. 2021; 1-9.

95.Van Der Gaag M, Smit F, Bechdolf A, French P, Linszen DH, Yung AR, et al. Preventing a first episode of psychosis: Meta-analysis of randomized controlled prevention trials of 12month and longer-term follow-ups. Schizophr Res. 2013; 149: 56-62.

96.Takizawa R, Fukuda M, Kawasaki S, Kasai K, Mimura M. NeuroImage Neuroimaging-aided differential diagnosis of the depressive state. Neuroimage. 2014; 85: 498-507.

97.Pruessner M, Cullen AE, Aas M, Walker EF. The neural diathesisstress model of schizophrenia revisited: An update on recent findings considering illness stage and neurobiological and methodological complexities. Neurosci Biobehav Rev. 2017; 73: 191-218.

98.Walker E, Mittal V, Tessner K. Stress and the hypothalamic pituitary adrenal axis in the developmental course of schizophrenia. Annu Rev Clin Psychol. 2008; 4: 189- 216.

99. Duncan-Johnson CC, Donchin E. The Relation of P300 Latency to Reaction Time as a Function of Expectancy *. Prog Brain Res. 1980; 54: 717-722.

100. Bassett DS, Bullmore E, Verchinski BA, Mattay VS, Weinberger DR, Meyer- Lindenberg A. Hierarchical organization of human cortical networks in health and Schizophrenia. J Neurosci. 2008; 28: 9239- 9248.

101. Van Den Heuvel MP, Sporns O, Collin G, Scheewe T, Mandl RCW, Cahn W, et al. Abnormal rich club organization and functional brain dynamics in schizophrenia. JAMA Psychiatry. 2013; 70: 783-792.

102. Zhang Y, Lin L, Lin CP, Zhou Y, Chou KH, Lo CY, et al. Abnormal topological organization of structural brain networks in schizophrenia. Schizophr Res. 2012; 141: 109-118.

103. Falkai P, Honer WG, Kamer T, Dustert S, Vogeley K, SchneiderAxmann T, et al. Disturbed frontal gyrification within families affected with schizophrenia. J Psychiatr Res. 2007; 41: 805-813.

104. Tepest R, Schwarzbach CJ, Krug B, Klosterko J, Ruhrmann S, Vogeley K. Morphometry of structural disconnectivity indicators in subjects at risk and in age- matched patients with schizophrenia. 2013; 15- 24.

105. Harris JM, Moorhead TWJ, Miller P, Mcintosh AM, Bonnici HM, Owens DGC, et al. Increased Prefrontal Gyrification in a Large High-Risk Cohort Characterizes Those Who Develop Prefrontal Development. 2007; 62: 722-729.

106. McGlashan TH, Addington J, Cannon T, Heinimaa M, McGorry P, O’Brien M, et al. Recruitment and treatment practices for helpseeking “prodromal” patients. Schizophr Bull. 2007; 33: 715-726.

107. Morrison AP, French P, Parker S, Roberts M, Stevens H, Bentall RP, et al. Three- year follow-up of a randomized controlled trial of cognitive therapy for the prevention of psychosis in people at ultrahigh risk. Schizophr Bull. 2007; 33: 682-687.

108. Singh S a NP. Outcome measures in early psychosis: relevance of duration of untreated psychosis. Br J Psychiatry. 2007; 191: 58-63

109. Kupfer DJ, Regier DA. Neuroscience, clinical evidence, and the future of psychiatric classification in DSM-5. Am J Psychiatry. 2011; 168: 672-674.

110. Wang L. Early diagnosis of breast cancer. Sensors (Switzerland). 2017; 17.

111. Kroll DS, Chakravartti A, Gasparrini K, Latham C, Davidson P, Byron-Burke M, et al. The walk-in clinic model improves access to psychiatry in primary care. J Psychosom Res. 2016; 89: 11-15.

112. Enderami A, Monesi F, Zarghami M. One-Year Follow-Up of Patients with a Diagnosis of First Episode Psychosis. Mater Socio Medica. 2017; 29: 21.

113. Kim JS, Baek JH, Choi JS, Lee D, Kwon JS, Hong KS. Diagnostic stability of first- episode psychosis and predictors of diagnostic shift from non-affective psychosis to bipolar disorder: A retrospective evaluation after recurrence. Psychiatry Res. 2011; 188: 29-33.

114. Guest FL, Guest PC, Martins-De-Souza D. The emergence of pointof-care blood- based biomarker testing for psychiatric disorders: enabling personalized medicine. Biomark Med. 2016; 10: 431-443.

115. Sakuma A, Obara C, Katsura M, Ito F, Ohmuro N, Iizuka K, et al. No regional gray matter volume reduction observed in young Japanese people at ultra-high risk for psychosis: A voxel-based morphometry study. Asian J Psychiatr. 2018; 37: 167-171.

116. Murphy VA, Shen MD, Kim SH, Cornea E, Styner M, Gilmore JH. Extraaxial Cerebrospinal Fluid Relationships to Infant Brain Structure, Cognitive Development, and Risk for Schizophrenia. Biol Psychiatry Cogn Neurosci Neuroimaging. 2020; 5: 651-659.

117. Da Silva T, Hafizi S, Rusjan PM, Houle S, Wilson AA, Prce I, et al. GABA levels and TSPO expression in people at clinical high risk for psychosis and healthy volunteers: A PET-MRS study. J Psychiatry Neurosci. 2019; 44: 111-1119.

118. Lizano P, Lutz O, Ling G, Lee AM, Eum S, Bishop JR, et al. Association of choroid plexus enlargement with cognitive, inflammatory, and structural phenotypes across the psychosis spectrum. Am J Psychiatry. 2019; 176: 564-572.

119. Soh P, Narayanan B, Khadka S, Calhoun VD, Keshavan MS, Tamminga CA, et al. Joint coupling of awake EEG frequency activity and MRI gray matter volumes in the psychosis dimension: a BSNIP study. Front psychiatry. 2015; 6: 162.

120. Hou J, Schmitt S, Meller T, Falkenberg I, Chen J, Wang J, et al. Cortical Complexity in People at Ultra-High-Risk for Psychosis Moderated by Childhood Trauma. Front Psychiatry. 2020; 11: 1-9.

121. Pelletier-Baldelli A, Orr JM, Bernard JA, Mittal VA. Social reward processing: A biomarker for predicting psychosis risk? Schizophr Res. 2020; 226: 129-137.

122. Hagenmuller F, Heekeren K, Meier M, Theodoridou A, Walitza S, Haker H, et al. The Loudness Dependence of Auditory Evoked Potentials (LDAEP) in individuals at risk for developing bipolar disorders and schizophrenia. Clin Neurophysiol. 2016; 127: 1342- 1350.

123. Siddiqui SV, Nizamie SH, Siddiqui MA, Jahan M, Garg S, Tikka SK, et al. Evaluation of N-400 Evoked Response Potential in schizophrenia: An endophenotype or a disease marker? Psychiatry Res. 2021; 300: 113907.

124. Koshiyama D, Kirihara K, Tada M, Nagai T, Fujioka M, Koike S, et al. Association between mismatch negativity and global functioning is specific to duration deviance in early stages of psychosis. Schizophr Res. 2018; 195: 378-384

125. Duffy FH, D’Angelo E, Rotenberg A, Gonzalez-Heydrich J. Neurophysiological differences between patients clinically at high risk for schizophrenia and neurotypical controls - first steps in development of a biomarker. BMC Med. 2015; 13.

126. Cruz G, Grent- T, Krishnadas R, Palva JM, Palva S, Uhlhaas PJ. Long range temporal correlations ( LRTCs) in MEG-data during emerging psychosis : Relationship to symptoms , medication-status and clinical trajectory. NeuroImage Clin. 2021; 31: 102722.

127. Tseng HH, Watts JJ, Kiang M, Suridjan I, Wilson AA, Houle S, et al. Nigral Stress- Induced Dopamine Release in Clinical High Risk and Antipsychotic-Naïve Schizophrenia. Schizophr Bull. 2018; 44: 542- 551.

128. Pollak TA, Kempton MJ, Iyegbe C, Vincent A, Irani SR, Coutinho E,et al. Clinical, cognitive and neuroanatomical associations of serum NMDAR autoantibodies in people at clinical high risk for psychosis. Mol Psychiatry. 2020; 26: 2590-2604.

129. Hafizi S, Da Silva T, Meyer JH, Kiang M, Houle S, Remington G, et al. Interaction between TSPO - A neuroimmune marker - And redox status in clinical high risk for psychosis: A PET-MRS study. Neuropsychopharmacology. 2018; 43: 1700-1705.

130. Obyedkov I, Skuhareuskaya M, Skugarevsky O, Obyedkov V, Buslauski P, Skuhareuskaya T, et al. Saccadic eye movements in different dimensions of schizophrenia and in clinical high-risk state for psychosis. BMC Psychiatry. 2019; 19: 1-10.

131. Schwab S, Jost M, Altorfer A. Impaired top-down modulation of saccadic latencies in patients with schizophrenia but not in firstdegree relatives. Front Behav Neurosci. 2015; 9: 1-7.

132. Söder E, Clamor A, Lincoln TM. Hair cortisol concentrations as an indicator of potential HPA axis hyperactivation in risk for psychosis. Schizophr Res. 2019; 212: 54-61

 

Painchaud A, Peredo R, Mérette C, Marquet P (2022) How can we Track Psychosis? A Scoping Review of Biomarkers of Transition in Subjects at Risk. JSM Biomar 5(1): 1014.

Received : 26 Apr 2022
Accepted : 24 May 2022
Published : 27 May 2022
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X