Abstract

Background and Hypothesis

This umbrella review aims to comprehensively synthesize the evidence of association between peripheral, electrophysiological, neuroimaging, neuropathological, and other biomarkers and diagnosis of psychotic disorders.

Study Design

We selected systematic reviews and meta-analyses of observational studies on diagnostic biomarkers for psychotic disorders, published until February 1, 2018. Data extraction was conducted according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines. Evidence of association between biomarkers and psychotic disorders was classified as convincing, highly suggestive, suggestive, weak, or non-significant, using a standardized classification. Quality analyses used the Assessment of Multiple Systematic Reviews (AMSTAR) tool.

Study Results

The umbrella review included 110 meta-analyses or systematic reviews corresponding to 3892 individual studies, 1478 biomarkers, and 392 210 participants. No factor showed a convincing level of evidence. Highly suggestive evidence was observed for transglutaminase autoantibodies levels (odds ratio [OR] = 7.32; 95% CI: 3.36, 15.94), mismatch negativity in auditory event-related potentials (standardized mean difference [SMD] = 0.73; 95% CI: 0.5, 0.96), P300 component latency (SMD = −0.6; 95% CI: −0.83, −0.38), ventricle-brain ratio (SMD = 0.61; 95% CI: 0.5, 0.71), and minor physical anomalies (SMD = 0.99; 95% CI: 0.64, 1.34). Suggestive evidence was observed for folate, malondialdehyde, brain-derived neurotrophic factor, homocysteine, P50 sensory gating (P50 S2/S1 ratio), frontal N-acetyl-aspartate, and high-frequency heart rate variability. Among the remaining biomarkers, weak evidence was found for 626 and a non-significant association for 833 factors.

Conclusions

While several biomarkers present highly suggestive or suggestive evidence of association with psychotic disorders, methodological biases, and underpowered studies call for future higher-quality research.

Introduction

Schizophrenia spectrum and other psychotic disorders have an estimated mean lifetime prevalence of 9.57 per 10001 and usually begin during youth and early adulthood (meta-analytic peak age of onset at 20.5 years2). The initial diagnosis of psychosis usually occurs at the time of the first episode of psychosis (FEP), occurring at a late stage in the neurodevelopmental trajectory of the disorder.3 Following a FEP, standard care with antipsychotics is primarily symptom-focused and limited for altering the course of the disorder.4 Psychotic disorders continue to be associated with poor clinical outcomes (1 in 7 recovery rate5) and a 3-fold excess mortality rate when compared to the general population (standardized mortality rate of 3.086) due to physical illness and increased suicide risk in the initial years following illness onset.7,8 Schizophrenia is a global leading cause of health-related disability,9 with associated global economic costs of up to 1.65% of the gross domestic product.10 Given the suboptimal results of current therapeutics, course-altering preventive interventions during the early developmental stages, such as cannabidiol11,12 or oxytocin,13,14 constitute promising future avenues for next-stage interventions.15 Course-altering interventions require a refined understanding of the pathophysiology and neurobiological substrates of psychosis.3 After decades of research, however, the causes of psychotic disorders remain elusive. The etiological models that have received the strongest empirical support suggest a complex combination of direct and interactive effects of genetic, epigenetic, and environmental factors across the developmental cycle that interfere with brain development and maturation.16,17

Nonetheless, the past decades have witnessed an explosion of psychosis biomarker research. Biomarkers provide clues for understanding the pathophysiological basis of psychosis,18 and could become key tools in the real-world implementation of precision medicine19,20 and individualized prediction modeling.21,22 For example, biomarkers can aid in developing mechanism-driven preventive interventions,23 identifying illness subtypes via biological screening,24 or in the introduction of novel diagnostic frameworks based on the pathophysiology of mental disorders.25 As such, biomarkers could be helpful for ascertaining the presence of a disorder or specific illness subtypes (“diagnostic” biomarkers), predicting therapeutic response (“predictive” biomarkers), and the course of the disorder (“prognostic” biomarkers), and for monitoring illness progression (“monitoring” biomarkers).26,27 However, the progress achieved in biomarker research has not been fully translated into real-world clinical practice,28 hence the current “translational gap.”29 Over 100 years following Bleuler’s introduction of the term “schizophrenia,”30 diagnosis is still based on clinical examination in accordance with DSM (Diagnostic and Statistical Manual of Mental Disorders) or ICD (International Classification of Diseases) diagnostic criteria.31 General psychopathological knowledge, and evidence-interventions, are implemented following a trial-and-error approach according to a general clinical profile.23,32

One obstacle for the translational potential of scientific findings is the fragmentation of knowledge as primary biomedical studies proliferate and diversify.33 This also affects secondary research due to the accumulation of overlapping, often contradictory, systematic reviews (SRs) and meta-analyses (MAs).34,35 Umbrella reviews help overcome this challenge by providing a synthesis and critical appraisal of SRs and MAs.36,37 For example, umbrella reviews for schizophrenia and psychotic disorders have focused on sociodemographic, developmental, and environmental risk factors,38 preventive treatments,15 or duration of untreated psychosis.39 Regarding biomarker research, umbrella reviews on schizophrenia and FEP have been limited to peripheral biomarkers, while also incorporating environmental exposures40 or other severe mental disorders.41 As such, to the best of our knowledge, no comprehensive umbrella review of biomarkers for psychosis has been published to date. Our study aims to close this gap by providing a state-of-the-art comprehensive evidence synthesis on the association between peripheral, electrophysiological, neuroimaging, neuropathological, and other biomarkers and psychotic disorders. We focus on observational studies of diagnostic biomarkers comparing individuals with psychosis vs healthy controls, as the analysis of prognostic markers would require a different design and evidence synthesis method which accounts for the time-dependency of outcomes. To increase sampling power, we use an extended sampling approach by including a diagnosis of any non-organic psychotic spectrum disorder, rather than limiting our selection to chronic and more symptomatic cases, such as schizophrenia.

In addition, biomarker research is often affected by underpowered samples, methodological biases, and inconsistent reporting practices.26,41 To address this challenge, we provide a hierarchical classification of the robustness of the association for each factor. To achieve this, we conduct a systematic analysis of biases through a set of a priori criteria extensively validated in previous risk factors studies for physical,42–48 neurological,49–52 and mental disorders,38,53–56 as well as in clinical studies.57–60 This classification into hierarchical levels of evidence is essential for reducing the ambiguities and contradictions often found in SRs and MAs.61

Methods

We pre-registered the umbrella review protocol with the International Prospective Register of Systematic Reviews (PROSPERO; CRD42017084377).

Search Strategy and Eligibility Criteria

Various researchers (PF-C and VR; plus JDA, EF, and CM for the neuroimaging part) systematically and independently searched PubMed, Web of Science, and Scopus through February 1, 2018, using the search terms (“systematic review” OR “meta-analysis”) and (“psychosis” OR “schizophrenia”), to identify SRs and MAs of studies examining potential diagnostic biomarkers for psychotic disorders. Reference lists of the SRs and MAs reaching full-text review were also carefully reviewed. Eligibility criteria included: (1) an SR or MA of individual observational studies examining associations between biological markers and psychotic disorders; (2) studies considering only established DSM or ICD diagnoses of non-organic psychotic spectrum disorders (eg, schizophrenia, schizoaffective, schizophreniform, affective psychosis [mania, depression, or bipolar disorder with psychosis], drug-induced psychosis, delusional disorder, brief psychotic disorder/acute, and transient psychotic disorder, psychosis not otherwise specified); (3) inclusion of a healthy control comparison group, and (4) studies reporting sufficient data to perform the analyses (or where data were retrievable from the authors). No language restrictions were applied.

When the biomarker dataset of an article was part of a larger dataset in another article, we only retained the latter. When two articles presented minimally overlapping datasets on the same biomarker, we used both SR or MA conjointly counting the overlapping primary studies only once. We excluded articles with an outcome other than established psychotic disorder, such as those related to relapse, remission, or treatment response of psychosis or symptom severity, and those investigating genetic markers for psychosis. We used the same inclusion/exclusion criteria for each study included in every eligible SR or MA.

Definition of Biomarker

We used the following accepted definition of biomarker: “a characteristic that is objectively measured and evaluated as an indicator of normal biological processes or pathogenic processes.”62 We did not include potential genetic biomarkers because umbrella reviews of genetic variables require different analytical methods and criteria.63 Neither did we include potential biomarkers from whole-brain voxel-based neuroimaging studies (although we did include other types of neuroimaging data) because we would need to treat each voxel as a biomarker. Instead, we refer the reader to existing MAs of whole-brain imaging studies in psychosis.64

We used the definition for each biomarker provided in the corresponding SR or MA. However, for reporting purposes, we classified biomarkers into the following categories: peripheral, electrophysiologic, neuropathological, neuroimaging, and other (eg, minor physical anomalies, high-frequency heart rate variability). The clustering of different biomarkers was pragmatically operationalized following the definition presented by each individual study. These groups hold only descriptive value as the actual analyses were performed at the single marker level. There was no assumption of biological mechanisms underlying these categories, most of which are yet to be fully elucidated.

Data Extraction and Selection

Data extraction was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA 2020) guidelines.65 Various investigators (PF-C and MAG-L; plus JDA, EF, CM, and AS for the neuroimaging part) conducted the following steps independently. First, we identified the potential biomarkers assessed in each selected SR or MA. Second, we confirmed that each article included in the SR or MA met our eligibility criteria for the umbrella review (ie, ICD/DSM diagnosis, healthy control group, and sufficient data for analysis). Third, we extracted the following data (from the SR or MA or, otherwise, from the individual study): (1) first author and year of publication, (2) the number of cases and controls, (3) effect size (ES) measure (standardized mean difference [SMD] for continuous biomarkers, odds ratio [OR] for binary biomarkers), and corresponding 95% confidence interval (CI), (4) means and standard deviations for cases and controls for continuous biomarkers, and the number of cases and controls with and without the biomarker for binary biomarkers. An exception was neuroimaging biomarkers, for which we only relied on the information reported in the MAs unless this information showed that the biomarker’s evidence could be stronger than class IV (see “Evidence stratification” section). An independent double extraction process was conducted for those biomarkers that presented unclear data. Fourth, for those biomarkers showing evidence of class I, II, or III, we rated the quality of the SRs or MAs that contributed studies for that biomarker using the Assessment of Multiple Systematic Reviews (AMSTAR) tool.66 Our quality ratings obtained a high interrater agreement (intraclass correlation = 0.924). For further information on the quality analysis, see Supplementary table 1.

Statistical Analyses

We conducted all analyses with the package “metaumbrella” for R (https://metaumbrella.org/),67 that performs all the calculations necessary for stratifying evidence in umbrella reviews.36,38,68–70 Specifically, it calculated, for each biomarker, the ES (Hedges’ g for continuous variables, OR for binary variables) and its confidence and prediction intervals, the between-study heterogeneity (I2 statistic), the Egger test to detect potential publication/reporting bias, and the excess significance bias test. Hedges’ g values represent SMDs between the patient and control groups and are interpreted as indicative of a “small” (g = 0.2), “medium” (g = 0.5), or “large” effect (g = 0.80).71 OR provides a measure of the likelihood of presenting any specific biomarker in cases vs healthy controls, with OR >1 indicating increased likelihood and OR <1 decreasing likelihood. The I2 statistic represents the percentage of total variance resulting from heterogeneity (ie, real differences in the studies’ ES), rather than chance. Egger’s test quantifies the relationship between sample size and ES, with significant results indicating the risk of publication bias.72 Finally, the excess significance bias test evaluates the relative presence of studies with excessive significant findings, by comparing the observed vs the expected number of studies with significant results.73

Evidence Stratification

We classified the strength of the evidence according to previous criteria38: class I (convincing) when the number of patients >1000, P < 10−6, I2 < 50%, the 95% prediction interval excludes the null, and no publication/reporting or excess significance biases are detected; class II (highly suggestive) when the evidence is weaker than convincing but the number of patients >1000, P < 10−6, and the largest study is statistically significant; class III (suggestive) when the evidence is weaker than highly suggestive but the number of patients >1000 and P < 10−3; and class IV (weak) when the evidence is lower than suggestive but P < .05.

Results

We included 110 SRs and MAs (Supplementary figure 1) covering 1478 potential biomarkers with a cumulative sample size of 189 180 individuals with psychosis and 203 030 healthy controls. In table 1 we include the biomarkers that achieved highly suggestive (class II) or suggestive (class III) level of evidence. For the full list of biomarkers, see Supplementary tables 2–6.

Table 1.

Biomarkers With Highly Suggestive or Suggestive Evidence of Association With Psychotic Disorders

FactorkDiagnosisES (95% CI)Features Used for Classification of Level of EvidenceeORCE
NCasesControlsPI2PI 95% CIEggerESBLS
Peripheral biomarkers
 tTG autoantibodies741SZOR, 7.32 (3.36, 15.94)230114019001 × 10−6NANANANoYes7.32II
 Folate7516SZSMD, −1.44 (−2.18, −0.71)2599126013391.20 × 10−498%−4.71, 1.83NoNoYes0.07III
 Malondialdehyde76–7826SZSMD, 1.38 (0.82, 1.94)179510777181 × 10−694%−1.56, 4.32NoYesYes12.22III
 BDNF79–8247FEP, SZSMD, −0.69 (−1.05, −0.33)4955275621992.00 × 10−491%−3.2, 1.82NoYesYes0.29III
 Homocysteine8319SZSMD, 0.6 (0.31, 0.89)3320130320175.1 × 10−589%−0.7, 1.91NoYesYes2.98III
Electrophysiologic biomarkers
 Mismatch negativity in auditory event-related potentials84–8747SZSMD, 0.73 (0.5, 0.96)564928712778<1 × 10−686%−0.81, 2.28NoYesYes3.77II
 P300 component latency88,8956SZSMD, −0.6 (−0.83, −0.38)350217351767<1 × 10−690%−2.22, 1.01NoYesYes0.33II
 P50 sensory gating (P50 S2/S1 ratio)88,90–9380AP, SZSMD, 0.79 (0.43, 1.16)4999210728922.0 × 10−593%−2.43, 4.02NoNoYes4.22III
Neuroimaging biomarkers
 Ventricle-brain ratio9472SZSMD, 0.61 (0.5, 0.71)609934632636<1 × 10−668%(−0.14, 1.35)NoYesYes3.00II
 Frontal NAA9568SZSMD, −0.34 (−0.47, −0.2)2868144414241 × 10−663%(−1.22, 0.55)NoYesNo0.54III
Other biomarkers
 Minor physical anomalies9614SZSMD, 0.99 (0.64, 1.34)216011531007<1 × 10−693%−0.45, 2.42NoYesYes5.99II
 High-frequency heart rate variability9728SZSMD, −0.99 (−1.41, −0.58)3008133016782 × 10−698%−3.24, 1.25YesYesYes0.16III
FactorkDiagnosisES (95% CI)Features Used for Classification of Level of EvidenceeORCE
NCasesControlsPI2PI 95% CIEggerESBLS
Peripheral biomarkers
 tTG autoantibodies741SZOR, 7.32 (3.36, 15.94)230114019001 × 10−6NANANANoYes7.32II
 Folate7516SZSMD, −1.44 (−2.18, −0.71)2599126013391.20 × 10−498%−4.71, 1.83NoNoYes0.07III
 Malondialdehyde76–7826SZSMD, 1.38 (0.82, 1.94)179510777181 × 10−694%−1.56, 4.32NoYesYes12.22III
 BDNF79–8247FEP, SZSMD, −0.69 (−1.05, −0.33)4955275621992.00 × 10−491%−3.2, 1.82NoYesYes0.29III
 Homocysteine8319SZSMD, 0.6 (0.31, 0.89)3320130320175.1 × 10−589%−0.7, 1.91NoYesYes2.98III
Electrophysiologic biomarkers
 Mismatch negativity in auditory event-related potentials84–8747SZSMD, 0.73 (0.5, 0.96)564928712778<1 × 10−686%−0.81, 2.28NoYesYes3.77II
 P300 component latency88,8956SZSMD, −0.6 (−0.83, −0.38)350217351767<1 × 10−690%−2.22, 1.01NoYesYes0.33II
 P50 sensory gating (P50 S2/S1 ratio)88,90–9380AP, SZSMD, 0.79 (0.43, 1.16)4999210728922.0 × 10−593%−2.43, 4.02NoNoYes4.22III
Neuroimaging biomarkers
 Ventricle-brain ratio9472SZSMD, 0.61 (0.5, 0.71)609934632636<1 × 10−668%(−0.14, 1.35)NoYesYes3.00II
 Frontal NAA9568SZSMD, −0.34 (−0.47, −0.2)2868144414241 × 10−663%(−1.22, 0.55)NoYesNo0.54III
Other biomarkers
 Minor physical anomalies9614SZSMD, 0.99 (0.64, 1.34)216011531007<1 × 10−693%−0.45, 2.42NoYesYes5.99II
 High-frequency heart rate variability9728SZSMD, −0.99 (−1.41, −0.58)3008133016782 × 10−698%−3.24, 1.25YesYesYes0.16III

Note: AP, affective psychosis; BDNF, brain-derived neurotrophic factor; CI, confidence interval; CE, class of evidence; eOR, equivalent odds ratio; Egger, significant Egger test; ES, effect size; ESB, excess significance bias; FEP, first episode of psychosis; k, number of studies for each factor; LS, largest study with significant effect; N, total number of participants; NA, not assessable; NAA, N-acetylaspartate; OR, odds ratio; PI, prediction interval; SMD, standardized mean difference; SZ, schizophrenia; tTG, transglutaminase.

Table 1.

Biomarkers With Highly Suggestive or Suggestive Evidence of Association With Psychotic Disorders

FactorkDiagnosisES (95% CI)Features Used for Classification of Level of EvidenceeORCE
NCasesControlsPI2PI 95% CIEggerESBLS
Peripheral biomarkers
 tTG autoantibodies741SZOR, 7.32 (3.36, 15.94)230114019001 × 10−6NANANANoYes7.32II
 Folate7516SZSMD, −1.44 (−2.18, −0.71)2599126013391.20 × 10−498%−4.71, 1.83NoNoYes0.07III
 Malondialdehyde76–7826SZSMD, 1.38 (0.82, 1.94)179510777181 × 10−694%−1.56, 4.32NoYesYes12.22III
 BDNF79–8247FEP, SZSMD, −0.69 (−1.05, −0.33)4955275621992.00 × 10−491%−3.2, 1.82NoYesYes0.29III
 Homocysteine8319SZSMD, 0.6 (0.31, 0.89)3320130320175.1 × 10−589%−0.7, 1.91NoYesYes2.98III
Electrophysiologic biomarkers
 Mismatch negativity in auditory event-related potentials84–8747SZSMD, 0.73 (0.5, 0.96)564928712778<1 × 10−686%−0.81, 2.28NoYesYes3.77II
 P300 component latency88,8956SZSMD, −0.6 (−0.83, −0.38)350217351767<1 × 10−690%−2.22, 1.01NoYesYes0.33II
 P50 sensory gating (P50 S2/S1 ratio)88,90–9380AP, SZSMD, 0.79 (0.43, 1.16)4999210728922.0 × 10−593%−2.43, 4.02NoNoYes4.22III
Neuroimaging biomarkers
 Ventricle-brain ratio9472SZSMD, 0.61 (0.5, 0.71)609934632636<1 × 10−668%(−0.14, 1.35)NoYesYes3.00II
 Frontal NAA9568SZSMD, −0.34 (−0.47, −0.2)2868144414241 × 10−663%(−1.22, 0.55)NoYesNo0.54III
Other biomarkers
 Minor physical anomalies9614SZSMD, 0.99 (0.64, 1.34)216011531007<1 × 10−693%−0.45, 2.42NoYesYes5.99II
 High-frequency heart rate variability9728SZSMD, −0.99 (−1.41, −0.58)3008133016782 × 10−698%−3.24, 1.25YesYesYes0.16III
FactorkDiagnosisES (95% CI)Features Used for Classification of Level of EvidenceeORCE
NCasesControlsPI2PI 95% CIEggerESBLS
Peripheral biomarkers
 tTG autoantibodies741SZOR, 7.32 (3.36, 15.94)230114019001 × 10−6NANANANoYes7.32II
 Folate7516SZSMD, −1.44 (−2.18, −0.71)2599126013391.20 × 10−498%−4.71, 1.83NoNoYes0.07III
 Malondialdehyde76–7826SZSMD, 1.38 (0.82, 1.94)179510777181 × 10−694%−1.56, 4.32NoYesYes12.22III
 BDNF79–8247FEP, SZSMD, −0.69 (−1.05, −0.33)4955275621992.00 × 10−491%−3.2, 1.82NoYesYes0.29III
 Homocysteine8319SZSMD, 0.6 (0.31, 0.89)3320130320175.1 × 10−589%−0.7, 1.91NoYesYes2.98III
Electrophysiologic biomarkers
 Mismatch negativity in auditory event-related potentials84–8747SZSMD, 0.73 (0.5, 0.96)564928712778<1 × 10−686%−0.81, 2.28NoYesYes3.77II
 P300 component latency88,8956SZSMD, −0.6 (−0.83, −0.38)350217351767<1 × 10−690%−2.22, 1.01NoYesYes0.33II
 P50 sensory gating (P50 S2/S1 ratio)88,90–9380AP, SZSMD, 0.79 (0.43, 1.16)4999210728922.0 × 10−593%−2.43, 4.02NoNoYes4.22III
Neuroimaging biomarkers
 Ventricle-brain ratio9472SZSMD, 0.61 (0.5, 0.71)609934632636<1 × 10−668%(−0.14, 1.35)NoYesYes3.00II
 Frontal NAA9568SZSMD, −0.34 (−0.47, −0.2)2868144414241 × 10−663%(−1.22, 0.55)NoYesNo0.54III
Other biomarkers
 Minor physical anomalies9614SZSMD, 0.99 (0.64, 1.34)216011531007<1 × 10−693%−0.45, 2.42NoYesYes5.99II
 High-frequency heart rate variability9728SZSMD, −0.99 (−1.41, −0.58)3008133016782 × 10−698%−3.24, 1.25YesYesYes0.16III

Note: AP, affective psychosis; BDNF, brain-derived neurotrophic factor; CI, confidence interval; CE, class of evidence; eOR, equivalent odds ratio; Egger, significant Egger test; ES, effect size; ESB, excess significance bias; FEP, first episode of psychosis; k, number of studies for each factor; LS, largest study with significant effect; N, total number of participants; NA, not assessable; NAA, N-acetylaspartate; OR, odds ratio; PI, prediction interval; SMD, standardized mean difference; SZ, schizophrenia; tTG, transglutaminase.

Peripheral Biomarkers

We included 284 peripheral biomarkers from 63 390 patients and 79 410 controls (cumulated sample sizes; Supplementary table 2). Transglutaminase (tTG) autoantibody levels achieved highly suggestive (class II) evidence with an OR = 7.32 (CI: 3.36, 15.94) (table 1; figure 1); we could not assess heterogeneity and potential biases because only 1 study was available for this analysis. Blood levels of folate, malondialdehyde (MDA), brain-derived neurotrophic factor (BDNF), and homocysteine (Hcy) showed class III (suggestive) evidence of association, with ESs of −1.44 (CI: −2.18, −0.71), 1.38 (CI: 0.82, 1.94), −0.69 (CI: −1.05, −0.33) and 0.60 (CI: 0. 31, 0.89), respectively. These class III biomarkers showed very large heterogeneity and (except for folate) potential excess significance bias. One hundred sixty-three other peripheral biomarkers achieved class IV evidence.

OR for tTG autoantibodies have been converted to equivalent Hedges’ g (eG) to allow for a comparison across risk factors. BDNF, brain-derived neurotrophic factor; CI, confidence interval; eG, equivalent Hedges’ g; SMD, standardized mean difference; tTG, transglutaminase.
Fig. 1.

Standardized mean differences for peripheral biomarkers and psychotic disorders.

Electrophysiologic Biomarkers

We examined 79 electrophysiologic biomarkers in 18 151 patients vs 19 346 controls (cumulated sample sizes; Supplementary table 3). Two biomarkers achieved class II evidence: mismatch negativity in auditory event-related potentials (ERP) (g = 0.73, CI: 0.50, 0.96) and P300 component latency (g = −0.60, CI: −0.83, −0.38) (table 1; figure 2). However, they showed very large heterogeneity and potential excess significance bias. On the other hand, P50 sensory gating achieved class III evidence with an ES of g = 0.79 (CI: 0.43, 1.16), but showed very large heterogeneity. Forty-one other biomarkers achieved class IV evidence.

CI, confidence interval; SMD, standardized mean difference.
Fig. 2.

Standardized mean differences for electrophysiological biomarkers and psychotic disorders.

Neuroimaging Biomarkers

We included 238 neuroimaging biomarkers in a cumulative sample size of 77 849 patients vs 73 184 controls (Supplementary table 4). Two of them showed class II or III evidence: ventricle-brain ratio (class II, g = 0.61, CI: 0.50, 0.71) and frontal N-acetylaspartate (NAA) levels (class III, g = −0.34, CI: −0.47, −0.20) (table 1; figure 3). The analyses suggested potential excess significance bias in both cases and the heterogeneity was large (>50%). One hundred forty-nine other neuroimaging biomarkers achieved class IV evidence.

CI, confidence interval; NAA, N-acetylaspartate; SMD, standardized mean difference.
Fig. 3.

Standardized mean differences for neuroimaging biomarkers and psychotic disorders.

Neuropathological Biomarkers

We studied a total of 406 neuropathological biomarkers in a cumulative sample size of 6170 patients vs 6526 controls (Supplementary table 5). One hundred thirteen biomarkers showed a statistically significant association with psychosis, all with class IV (weak) evidence.

Other Biomarkers

The other biomarkers category enclosed 471 biomarkers in 23 620 patients vs 24 564 controls (cumulated sample sizes; Supplementary table 6). Minor physical anomalies achieved class II evidence (g = 0.99, CI: 0.64, 1.34) (table 1; figure 4), although with very large heterogeneity and potential excess significance bias. High-frequency heart rate variability achieved class III evidence (g = −0.99, CI: −1.41, −0.58), although, beyond very large heterogeneity, it also showed potential publication and excess significance biases. One hundred sixty other biomarkers achieved class IV evidence.

CI, confidence interval; SMD, standardized mean difference.
Fig. 4.

Standardized mean differences for other biomarkers and psychotic disorders.

Discussion

To the best of our knowledge, this is the first comprehensive umbrella review of diagnostic biomarkers for psychosis that incorporates peripheral, electrophysiologic, neuroimaging, neuropathological, and other biomarkers, in addition to a robust hierarchical classification of evidence. Overall, 110 SRs and MAs, a total of 3892 individual studies and 392 210 participants, and 1478 potential biomarkers were included. None of the evaluated biomarkers presented convincing evidence (class I) of association. Nonetheless, there was highly suggestive evidence (class II) for 5 biomarkers (0.3% of all evaluated factors), and suggestive evidence (class III) for 7 biomarkers (0.5%). Among the remaining biomarkers, 626 (42.6%) presented significant but weak (class IV) evidence. The remaining 833 (56.6%) presented non-significant associations.

Associations supported by highly suggestive and suggestive evidence merit discussion. Regarding peripheral biomarkers, highly suggestive evidence was only found for elevated levels of tTG autoantibodies. This finding provides evidence, for a subset of individuals, towards a link between psychosis and celiac disease.98,99 The relationship between celiac disease or gluten sensitivity and neurologic and psychiatric disorders was initially reported almost 70 years ago,100,101 with celiac disease having since been linked to an increased risk of various neurological102 and mental disorders.103 In addition, it provides evidence for psychosis as a systemic disorder with potential autoimmune components.104,105 While the specific nature of the association remains unknown, possible mechanisms include shared genetic susceptibility or immunological abnormalities.106 However, the estimate is based on a single study. As a result, heterogeneity or publication bias could not be assessed. Nonetheless, the large sample of cases (n = 2301) grants this association a class II level of evidence. In addition, we found suggestive evidence for reduced blood levels of folate among individuals with psychosis, although this association was characterized by very large heterogeneity. This finding further indicates potential nutrition-related mechanisms in mental disorders.107–109 For example, maternal exposure to nutritional deprivation during pregnancy110 could result in deficits in micronutrients involved in 1-carbon metabolism, particularly folate (vitamin B9) and vitamin B12, leading to increased risk due to epigenetic changes via disruptions in DNA methylation.111–113 Findings of decreased levels of vitamin B12 have been inconsistent114,115 and additional epidemiological and laboratory studies are required. We also acknowledge that there might be a bi-directional effect of psychosis on diet116 and therefore no causal assumption is made when interpreting diet-related biomarkers. The scarcity of prospective studies is problematic, as folate and vitamin B12 levels could be confounded by the use of antipsychotics.117 We also observed suggestive evidence for increased plasma levels of Hcy, further indicating a potential role of 1-carbon metabolism. Hcy is a non-protein amino acid produced in 1-carbon methyl group-transfer metabolism with various functions in brain activity,118 and its regulation depends on dietary folate and other B vitamins.83 In schizophrenia patients, plasma Hcy levels were found to correlate negatively with folate119 and vitamin B12,120 and to be associated with symptom severity118,121,122 and a progressive course of illness.123 Elevated Hcy has been observed in FEP patients,124,125 although not in clinical high-risk for psychosis (CHR-P) individuals.126 However, research in the CHR-P population is still in an emerging state. Potential mechanisms involved in the association between Hcy and psychosis include aberrant DNA methylation, altered NMDA receptor and glutamatergic transmission, toxic effects on dopaminergic neurons, premature apoptosis, oxidative stress, and placental vascular damage and fetal hypoxia.118–120,127,128 Nonetheless, the question remains as to whether hyperhomocysteinemia is a contributor, a consequence, or an epiphenomenon of psychosis.120 Adjunct folate therapy has been found to improve symptoms of depressive and bipolar disorders, but not for schizophrenia.129 However, emerging evidence suggests moderate effectiveness of pooled vitamin B (ie, B6, B9, B12) supplementation on total schizophrenia symptoms.130 It is still unclear what specific symptoms and clinical groups (eg, FEP, chronic) would benefit the most from nutritional interventions, and an opportunity for clinical stratification might exist.4 For example, folic acid appears to be more beneficial for the treatment of negative vs positive symptoms.131 Likewise, a recent randomised clinical trial (RCT) study suggested that B-vitamin supplementation could have neuroprotective effects in FEP patients with elevated Hcy.132 Also, it is unclear how nutritional deficiencies might interact with genetic variants.83,133 For example, genetic variants linked to folate-metabolism can be associated with clinical response to adjunct folate therapy.134,135 Overall, these findings call for further research into gene-environment interaction17 and potential avenues for personalized medicine.

Finally, regarding peripheral biomarkers, elevated levels of MDA and reduced levels of BDNF also presented suggestive evidence which, despite large heterogeneity, was based on large samples (n = 1795 and n = 4955, respectively). Alongside increased Hcy, Elevated MDA and reduced BDNF levels suggest a role for oxidative stress and inflammation in psychosis pathophysiology, possibly linked to immune dysregulation.136 Immune dysregulation in psychosis has been supported by genome-wide association137,138 and postmortem studies.139 Early-life adversity, including prenatal insults110 and childhood trauma,38 can also promote a pro-inflammatory state later in life, potentially via epigenetic changes.140,141 We also observed weak evidence for other oxidative stress and inflammation biomarkers, such as docosahexaenoic acid (DHA), IL-6, TNF-alpha, and total antioxidant status. Overall, these findings are consistent with evidence suggestive of antioxidant status and pro-inflammatory imbalances among FEP patients125 and CHR-P individuals.142 However, these associations are often weakened by small samples, scarcity of prospective studies, and high heterogeneity potentially resulting from between-study sample differences (eg, illness phase, developmental stage, antipsychotic status, and comorbid substance-user or other lifestyle factors).76,125 Heterogeneity and small ESs might further indicate that immune system involvement varies between individuals and across illness phases.143 For example, anti-inflammatory agents have been found to be more effective for early-phase vs chronic schizophrenia.144 Also, anti-inflammatory biomarkers can be associated with antipsychotic treatment response.145 As such, an opportunity might also exist for personalized medicine approaches22 via inflammatory biomarker-informed patient stratification.

In terms of electrophysiologic biomarkers, mismatch negativity (MMN) in auditory ERP achieved highly suggestive evidence. Reduced MMN is a well-replicated feature in first-episode and chronic psychosis,28,84,85,146 and might reflect N-methyl-d-aspartate (NMDA) receptor function.147,148 MMN does not seem to always distinguish between early and chronic schizophrenia,149,150 and has been suggested to stabilize following deterioration during the first 1–2 years of diagnosis.146 In addition, abnormal auditory MMN has also been observed to precede illness onset among CHR-P individuals,151–153 although not consistently,150 and with other studies reporting similar patterns in CHR-P and FEP individuals.154 Overall, there is translational potential for MMN as a biomarker for monitoring illness progression and the identification of individuals at greater risk of transition.146,155 In addition, we observed highly suggestive evidence for P300 component latency. Significant evidence, albeit weak, was also observed for P300 amplitude disturbances. Both P300 component latency and amplitude, a more direct biomarker of cognition than MMN,156–158 have been widely replicated in first-episode159–161 and chronic psychosis patients,88,162 and first-degree relatives.163 P300 anomalies have also been reported to respond to antipsychotic treatment in schizophrenia patients,164 and might be linked to gray matter volume reduction in both CHR-P165,166 and psychosis samples.161 As such, ERP deficits have the potential as biomarkers for transition risk in clinical high-risk individuals160,167–169 in whom both neurocognitive decline170 and reduction in gray matter volume171 precede illness onset. Finally, P50 sensory gating (P50 S2/S1 ratio) presented suggestive evidence, further supporting the existence of ERP deficits in psychosis.28 All class II and class III electrophysiologic biomarkers were affected by large heterogeneity.

Among neuroimaging biomarkers, we observed highly suggestive evidence of increased ventricle-brain ratio among individuals with psychosis. Since the pioneer neuroimaging studies of the 1970–1980’s,172–175 ventricular enlargement has remained one of the most replicated neuroanatomical correlated of schizophrenia, as reflected in the large number of participants in our summary (n = 6099). Ventricular enlargement has also been observed in unaffected first-degree relatives176 and in transitioning CHR-P samples.177 In addition, decreased frontal NAA achieved the suggestive level of evidence. Weak evidence for reduced NAA was also present for other brain regions (eg, temporal, cerebellum, and thalamus), further supporting an association between psychosis and loss of neuronal integrity.3 Deterioration in neuronal health might precede illness onset, as suggested by evidence for CHR-P individuals,95,178 although findings in this group have been inconsistent179 and better-powered prospective studies are required. Overall, neuroimaging evidence is indicative of aberrant brain trajectories that seem to precede illness onset,180,181 suggesting a neurodevelopmental component to the disorder,16 and that have potential as markers for transition risk in high-risk individuals.182 Following illness onset, comorbid substance use, antipsychotic medication, stress, and lifestyle factors can contribute to progressive ventricular volume increases,183,184 in addition to underlying neurobiological factors. As such, high between-study heterogeneity observed in neuroimaging biomarkers might reflect methodological differences affecting the selection of controls, diagnostic criteria, and clinical characteristics of included cases.185

For neuropathological biomarkers, none of the 406 assessed factors obtained highly suggestive or suggestive evidence. While 113 biomarkers did present significant associations, the level of evidence was weak, with most (95.6%) estimates being based on a single study, and all of them employing small samples (range of n = 9–176).

Regarding other biomarkers, minor physical abnormalities presented highly suggestive evidence, although very large heterogeneity and potential excess significance bias were observed. This finding is consistent with extensive evidence of early-life neurodevelopmental deviance in psychosis, including an excess of prenatal and perinatal insults,110 childhood motor186 and cognitive187 abnormalities, and structural brain alterations.181,188,189 Originally, these observations led to the neurodevelopmental hypothesis of schizophrenia,190,191 later expanded to incorporate environmental adversity occurring during childhood and adolescence.16,192 Finally, lower high-frequency heart rate variability achieved suggestive evidence. However, this association is affected by large heterogeneity potential publication, and excess significance bias. Nonetheless, this finding supports the hypothesis of reduced vagal activity among individuals with psychotic disorders as a potential endophenotype associated with executive function,193 emotional regulation,194,195 and threat inhibition.196 This is also consistent with the evidence of an increased risk of cardiovascular disease among individuals with schizophrenia.197 While it is unclear how autonomic function alterations manifest in CHR-P individuals,198,199 an increase in high-frequency heart rate variability has been observed in CHR-P men following intranasal oxytocin,14 suggesting possible novel disease-engagement psychopharmacological targets.

Overall, the evidence summarized in this study presents some limitations. First, we provide estimates of association and do not assume a cause-effect relationship between psychotic disorders and the reported biomarkers. Reverse causality200 is an important component in biomarker research, as the temporality of the exposure cannot always be ascertained. Furthermore, we found common methodological limitations in psychosis biomarker research. On one hand, large statistical heterogeneity (I2 > 50%) was observed in 12.1% of all 1478 factors, and in 91.7% of factors with class II or III evidence. In addition, between-study heterogeneity could not be evaluated for 74.5% of biomarkers, and for 1 factor with class II evidence. This could indicate between-study methodological differences related to data collection and reporting, or sampling criteria, which can limit the reproducibility and real-world translation of findings. High heterogeneity can also be indicative of real differences between clinical or biological subtypes, meriting further exploration.24,201 In addition to high heterogeneity, excess significance bias (affecting 75% of class II and III biomarkers) and the limited number of cases of <1000 (affecting 92.4% of all 1478 factors) were frequently observed. Inflated ESs have been suggested to particularly affect newly discovered associations due to suboptimal power.202 In our sample, 38.8% of all factors based on a single study reported statistically significant associations. Among 865 estimates based on a single study (affecting 58.5% of all factors), all but one had a sample of cases of <1000 participants. High heterogeneity, excess significance bias, and small samples are methodological limitations not restrictive of psychosis research and have also been observed in biomarker research for other mental disorders.41,68,203,204 Furthermore, reporting bias via the Egger test could not be assessed for 87.5% of all factors due to insufficient data or due to only one study being included, and was significant for 1.1% and non-significant for 11.4% of biomarkers. Overall, these results highlight the need for better-powered replication studies with representative samples that will allow us to assess whether these biomarkers are generalizable to other settings with different sociodemographics and service configurations. Without this understanding, the prospects of implementing these biomarkers in real-world clinical care and benefitting patients are severely limited.205 Moreover, implementing a biomarker that is only useful in certain (usually majority) sub-populations or settings can lead to harm and perpetuation of existing health inequities.206,207 Ongoing large multisite research networks are being conducted to address these limitations.208,209 In addition, compliance with common guidelines and practices for evaluating and reporting biomarkers210 is needed to increase the replicability of findings and minimize missing data. Another limitation is that the current literature search is not fully updated. This is due to the high complexity of data extraction, data quality check procedures, and synthesis that are needed for this type of analysis. However, our robust method could be leveraged by subsequent evidence syntheses in this field to mainstream updated summaries. As a final limitation of our report, we did not assess the quality of individual primary studies included in the evidence synthesis as this was outside the scope of our review. In the same lines, the scope of this review was limited to diagnostic biomarkers and we did not assess prognostic biomarkers or those useful to forecast drug response.211

Conclusion

Several biomarkers presented highly suggestive or suggestive evidence of association with psychotic disorders, indicating various translational opportunities for personalized medicine. However, the widespread presence of methodological biases and underpowered studies in biomarker research highlight the need for future higher-quality research.

Acknowledgments

GA has received CME-related honoraria, or consulting fees from Adamed, Angelini, Casen Recordati, Janssen-Cilag, Lundbeck, Lundbeck/Otsuka, Rovi, and Viatris, with no financial or other relationship relevant to the subject of this article. EV has received grants and served as a consultant, advisor, or CME speaker for the following entities: AB-Biotics, AbbVie, Angelini, Biogen, Boehringer Ingelheim, Celon Pharma, Dainippon Sumitomo Pharma, Ferrer, Gedeon Richter, GH Research, Glaxo-Smith Kline, Janssen, Lundbeck, Novartis, Orion Corporation, Organon, Otsuka, Sage, Sanofi-Aventis, Sunovion, Takeda, and Viatris, outside the submitted work. JR has received CME honoraria from Inspira Networks for a machine learning course promoted by Adamed, outside the submitted work. PFP has received research funds or personal fees from Lundbeck, Angelini, Menarini, Sunovion, Boehringer Ingelheim, Mindstrong, and Proxymm Science, outside the current study.

Funding

This work was funded by a Wellcome Trust grant to PFP (Early DetectioN of menTal disorERs, ENTER: 215793/Z/19/Z) and the Catalonian Government (2017SGR01271 and 2021SGR01475 to EP-C), CIBERSAM and Instituto de Salud Carlos III, co-funded by European Union (FEDER/FSE, “Investing in your future”): Sara Borrell contract (CD22/00106 to MAG-L), Research Project Grant (PI21/00416 to EP-C). PF-C is funded by a fellowship from “la Caixa” Foundation (ID 100010434, fellowship LCB/BQ/PR22/11920017). MAG-L was also funded by a Júlia Gil Pineda Research Fellowship. GA is supported by a Juan Rodés 2023 grant (JR23/00050), a Rio Hortega 2021 grant (CM21/00017), and an Acción Estratégica en Salud - Mobility (M-AES) fellowship (MV22/00058), from the Spanish Ministry of Health, financed by the Instituto de Salud Carlos III (ISCIII) and co-financed by the Fondo Social Europeo Plus (FSE+). JR’s work is supported by the Spanish Ministry of Science and Innovation (PI19/00394 and PI22/00261), integrated into the Plan Nacional de I+D+I and co-financed by ERDF Funds from the European Commission (“A Way of Making Europe”) and the CERCA Program/Generalitat de Catalunya and Secretaria d’Universitats i Recerca del Departament d’Economia I Coneixement (2021 SGR 01128).

References

1.

Moreno-Küstner
B
,
Martín
C
,
Pastor
L.
Prevalence of psychotic disorders and its association with methodological issues. A systematic review and meta-analyses
.
PLoS One.
2018
;
13
(
4
):
e0195687
e0195625
.

2.

Solmi
M
,
Radua
J
,
Miriam
O
, et al.
Age at onset of mental disorders worldwide: a large-scale meta-analysis of 192 epidemiological studies
.
Mol Psychiatry.
2022
;
27
:
281
295
.

3.

Millan
MJ
,
Andrieux
A
,
Bartzokis
G
, et al.
Altering the course of schizophrenia: progress and perspectives
.
Nat Rev Drug Discov.
2016
;
15
(
7
):
485
515
.

4.

Fusar-Poli
P
,
McGorry
PD
,
Kane
JM.
Improving outcomes of first-episode psychosis: an overview
.
World Psychiatry.
2017
;
16
(
3
):
251
265
.

5.

Jääskeläinen
E
,
Juola
P
,
Hirvonen
N
, et al.
A systematic review and meta-analysis of recovery in schizophrenia
.
Schizophr Bull.
2013
;
39
(
6
):
1296
1306
.

6.

Oakley
P
,
Kisely
S
,
Baxter
A
, et al.
Increased mortality among people with schizophrenia and other non-affective psychotic disorders in the community: a systematic review and meta-analysis
.
J Psychiatr Res.
2018
;
102
:
245
253
.

7.

De Hert
M
,
Correll
C
,
Bobes
J
, et al.
Physical illness in patients with severe mental disorders. I. Prevalence, impact of medications and disparities in health care
.
World Psychiatry.
2011
;
10
(
1
):
52
77
.

8.

Hjorthøj
C
,
Stürup
AE
,
McGrath
JJ
,
Nordentoft
M.
Years of potential life lost and life expectancy in schizophrenia: a systematic review and meta-analysis
.
Lancet Psychiatry.
2017
;
4
(
4
):
295
301
.

9.

Global Burden of Disease 2017 Disease and Injury Incidence and Prevalence Collaborators
.
Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016
.
Lancet.
2017
;
390
:
1211
1259
.

10.

Chong
HY
,
Teoh
SL
,
Wu
DBC
,
Kotirum
S
,
Chiou
CF
,
Chaiyakunapruk
N.
Global economic burden of schizophrenia: a systematic review
.
Neuropsychiatr Dis Treat.
2016
;
12
:
357
373
.

11.

Amminger
GP
,
Lin
A
,
Kerr
M
, et al.
Cannabidiol for at risk for psychosis youth: a randomized controlled trial
.
Early Interv Psychiatry.
2022
;
16
(
4
):
419
432
.

12.

Chesney
E
,
Oliver
D
,
McGuire
P.
Cannabidiol (CBD) as a novel treatment in the early phases of psychosis
.
Psychopharmacology (Berl).
2022
;
239
(
5
):
1179
1190
.

13.

Davies
C
,
Paloyelis
Y
,
Rutigliano
G
, et al.
Oxytocin modulates hippocampal perfusion in people at clinical high risk for psychosis
.
Neuropsychopharmacology.
2019
;
44
(
7
):
1300
1309
.

14.

Martins
D
,
Davies
C
,
Micheli
AD
,
Oliver
D
,
Fusar-Poli
P
,
Paloyelis
Y.
Intranasal oxytocin increases heart-rate variability in men at clinical high risk for psychosis: a proof-of-concept study
.
Transl Psychiatry.
2020
;
10
(
227
):
1
12
.

15.

Fusar-Poli
P
,
Davies
C
,
Solmi
M
, et al.
Preventive treatments for psychosis: umbrella review (just the evidence)
.
Front Psychiatry.
2019
;
10
:
1
21
.

16.

Howes
OD
,
Murray
RM.
Schizophrenia: an integrated sociodevelopmental-cognitive model
.
Lancet.
2014
;
383
:
1677
1687
.

17.

Van Os
J
,
Rutten
BPF
,
Poulton
R.
Gene-environment interactions in schizophrenia: review of epidemiological findings and future directions
.
Schizophr Bull.
2008
;
34
(
6
):
1066
1082
.

18.

Chana
G
,
Bousman
CA
,
Money
TT
, et al.
Biomarker investigations related to pathophysiological pathways in schizophrenia and psychosis
.
Front Cell Neurosci.
2013
;
7
:
1
18
.

19.

Vieta
E.
La medicina personalizada aplicada a la salud mental: la psiquiatría de precisión
.
Rev Psiquiatr Salud Ment.
2015
;
8
(
2
):
117
118
.

20.

Salagre
E
,
Vieta
E.
Precision psychiatry: complex problems require complex solutions
.
Eur Neuropsychopharmacol.
2021
;
52
:
94
95
.

21.

Fusar-Poli
P
,
Correll
CU
,
Arango
C
,
Berk
M
,
Patel
V
,
Ioannidis
J.
Preventive psychiatry: a blueprint for improving the mental health of young people
.
World Psychiatry.
2021
;
20
(
2
):
200
221
.

22.

Fraguas
D
,
State
MW
,
Gur
RE
, et al.
Mental disorders of known aetiology and precision medicine in psychiatry: a promising but neglected alliance
.
Psychol Med.
2017
;
47
(
2
):
193
197
.

23.

Arango
C
,
Kapur
S
,
Kahn
RS.
Going beyond “trial-and-error” in psychiatric treatments: OPTiMiSE-ing the treatment of first episode of schizophrenia
.
Schizophr Bull.
2015
;
41
(
3
):
546
548
.

24.

Kapur
S
,
Phillips
AG
,
Insel
TR.
Why has it taken so long for biological psychiatry to develop clinical tests and what to do about it
?
Mol Psychiatry.
2012
;
17
:
1174
1179
.

25.

Insel
TR
,
Cuthbert
B
,
Garvey
M
, et al.
Research Domain Criteria (RDoC): toward a new classification framework for research on mental disorders
.
Am J Psychiatry.
2010
;
167
(
7
):
748
751
.

26.

Prata
D
,
Mechelli
A
,
Kapur
S.
Clinically meaningful biomarkers for psychosis: a systematic and quantitative review
.
Neurosci Biobehav Rev.
2014
;
45
:
134
141
.

27.

Peterson
BS.
Editorial: Biomarkers in precision medicine for mental illnesses
.
J Child Psychol Psychiatry.
2020
;
61
(
12
):
1279
1281
.

28.

Lawrie
SM
,
Olabi
B
,
Hall
J
,
Mcintosh
AM.
Do we have any solid evidence of clinical utility about the pathophysiology of schizophrenia
?
World Psychiatry.
2011
;
10
(
1
):
19
31
.

29.

Fernandes
BS
,
Williams
LM
,
Steiner
J
,
Leboyer
M
,
Carvalho
AF
,
Berk
M.
The new field of “precision psychiatry”
.
BMC Med.
2017
;
15
(
1
):
1
7
.

30.

Berrios
GE.
Eugen Bleuler’s place in the history of psychiatry
.
Schizophr Bull.
2011
;
37
(
6
):
1095
1098
.

31.

Stein
DJ
,
Szatmari
P
,
Gaebel
W
, et al.
Mental, behavioral and neurodevelopmental disorders in the ICD-11: an international perspective on key changes and controversies
.
BMC Med.
2020
;
18
(
21
):
1
24
.

32.

Leucht
S
,
Hierl
S
,
Kissling
W
,
Dold
M
,
Davis
JM.
Putting the efficacy of psychiatric and general medicine medication into perspective: review of meta-analyses
.
Br J Psychiatry.
2012
;
200
(
2
):
97
106
.

33.

Ioannidis
JPA.
Evolution and translation of research findings: from bench to where
?
PLoS Clin Trials.
2006
;
1
(
7
):
e36
e15
.

34.

Siontis
KC
,
Hernandez-Boussard
T
,
Ioannidis
JPA.
Overlapping meta-analyses on the same topic: survey of published studies
.
BMJ.
2013
;
347
:
f4501
.

35.

Ioannidis
JPA.
The mass production of redundant, misleading, and conflicted systematic reviews and meta-analyses
.
Milbank Q.
2016
;
94
(
3
):
485
514
.

36.

Fusar-Poli
P
,
Radua
J.
Ten simple rules for conducting umbrella reviews
.
Evid Based Ment Health.
2018
;
21
(
3
):
95
100
.

37.

Ioannidis
JPA.
Integration of evidence from multiple meta-analyses: a primer on umbrella reviews, treatment networks and multiple treatments meta-analyses
.
CMAJ.
2009
;
181
(
8
):
488
493
.

38.

Radua
J
,
Ramella-Cravaro
V
,
Ioannidis
JPA
, et al.
What causes psychosis? An umbrella review of risk and protective factors
.
World Psychiatry.
2018
;
17
(
1
):
49
66
.

39.

Howes
OD
,
Whitehurst
T
,
Shatalina
E
, et al.
The clinical significance of duration of untreated psychosis: an umbrella review and random-effects meta-analysis
.
World Psychiatry.
2021
;
20
(
1
):
75
95
.

40.

Belbasis
L
,
Köhler
CA
,
Stefanis
N
, et al.
Risk factors and peripheral biomarkers for schizophrenia spectrum disorders: an umbrella review of meta-analyses
.
Acta Psychiatr Scand.
2018
;
137
(
2
):
88
97
.

41.

Carvalho
AF
,
Solmi
M
,
Sanches
M
, et al.
Evidence-based umbrella review of 162 peripheral biomarkers for major mental disorders
.
Transl Psychiatry.
2020
;
10
(
1
):
1
13
.

42.

Dinu
M
,
Pagliai
G
,
Casini
A
,
Sofi
F.
Mediterranean diet and multiple health outcomes: an umbrella review of meta-analyses of observational studies and randomised trials
.
Eur J Clin Nutr.
2018
;
72
(
1
):
30
43
.

43.

Markozannes
G
,
Tzoulaki
I
,
Karli
D
, et al.
Diet, body size, physical activity and risk of prostate cancer: an umbrella review of the evidence
.
Eur J Cancer.
2016
;
69
:
61
69
.

44.

Belbasis
L
,
Savvidou
MD
,
Kanu
C
,
Evangelou
E
,
Tzoulaki
I.
Birth weight in relation to health and disease in later life: an umbrella review of systematic reviews and meta-analyses
.
BMC Med.
2016
;
14
(
1
):
147
.

45.

Belbasis
L
,
Stefanaki
I
,
Stratigos
AJ
,
Evangelou
E.
Non-genetic risk factors for cutaneous melanoma and keratinocyte skin cancers: an umbrella review of meta-analyses
.
J Dermatol Sci.
2016
;
84
(
3
):
330
339
.

46.

Kyrgiou
M
,
Kalliala
I
,
Markozannes
G
, et al.
Adiposity and cancer at major anatomical sites: umbrella review of the literature
.
BMJ.
2017
;
356
:
j410
j477
.

47.

Tsilidis
KK
,
Kasimis
JC
,
Lopez
DS
,
Ntzani
EE
,
Ioannidis
JPA.
Type 2 diabetes and cancer: umbrella review of meta-analyses of observational studies
.
BMJ.
2015
;
350
:
1
11
.

48.

Ioannidis
JPA
,
Zhou
Y
,
Chang
CQ
,
Schully
SD
,
Khoury
MJ
,
Freedman
AN.
Potential increased risk of cancer from commonly used medications: an umbrella review of meta-analyses
.
Ann Oncol.
2014
;
25
(
1
):
16
23
.

49.

Bellou
V
,
Belbasis
L
,
Tzoulaki
I
,
Middleton
LT
,
Ioannidis
JPA
,
Evangelou
E.
Systematic evaluation of the associations between environmental risk factors and dementia: an umbrella review of systematic reviews and meta-analyses
.
Alzheimers Dement.
2017
;
13
(
4
):
406
418
.

50.

Bellou
V
,
Belbasis
L
,
Tzoulaki
I
,
Evangelou
E
,
Ioannidis
JPA.
Environmental risk factors and Parkinson’s disease: an umbrella review of meta-analyses
.
Parkinsonism Relat Disord.
2016
;
23
:
1
9
.

51.

Belbasis
L
,
Bellou
V
,
Evangelou
E.
Environmental risk factors and amyotrophic lateral sclerosis: an umbrella review and critical assessment of current evidence from systematic reviews and meta-analyses of observational studies
.
Neuroepidemiology.
2016
;
46
(
2
):
96
105
.

52.

Belbasis
L
,
Bellou
V
,
Evangelou
E
,
Ioannidis
JPA
,
Tzoulaki
I.
Environmental risk factors and multiple sclerosis: an umbrella review of systematic reviews and meta-analyses
.
Lancet Neurol.
2015
;
14
(
3
):
263
273
.

53.

Solmi
M
,
Dragioti
E
,
Arango
C
, et al.
Risk and protective factors for mental disorders with onset in childhood/adolescence: an umbrella review of published meta-analyses of observational longitudinal studies
.
Neurosci Biobehav Rev.
2021
;
120
:
565
573
.

54.

Arango
C
,
Dragioti
E
,
Solmi
M
, et al.
Risk and protective factors for mental disorders beyond genetics: an evidence-based atlas
.
World Psychiatry.
2021
;
20
(
3
):
417
436
.

55.

Bortolato
B
,
Köhler
CA
,
Evangelou
E
, et al.
Systematic assessment of environmental risk factors for bipolar disorder: an umbrella review of systematic reviews and meta-analyses
.
Bipolar Disord.
2017
;
19
(
2
):
84
96
.

56.

Kim
JH
,
Kim
JY
,
Lee
J
, et al.
Environmental risk factors, protective factors, and peripheral biomarkers for ADHD: an umbrella review
.
Lancet Psychiatry.
2020
;
7
(
11
):
955
970
.

57.

Dragioti
E
,
Karathanos
V
,
Gerdle
B
,
Evangelou
E.
Does psychotherapy work? An umbrella review of meta-analyses of randomized controlled trials
.
Acta Psychiatr Scand.
2017
;
136
(
3
):
236
246
.

58.

Papageorgiou
P
,
Deschner
J
,
Papageorgiou
S.
Effectiveness and adverse effects of deep brain stimulation: umbrella review of meta-analyses
.
J Neurol Surg.
2016
;
78
(
2
):
180
190
.

59.

Tonelli
A
,
Zein
J
,
Adams
J
,
Ioannidis
J.
Effects of interventions on survival in acute respiratory distress syndrome: an umbrella review of 159 published randomized trials and 29 meta-analyses
.
Intensive Care Med.
2014
;
40
(
6
):
769
787
.

60.

Barbui
C
,
Purgato
M
,
Abdulmalik
J
, et al.
Efficacy of psychosocial interventions for mental health outcomes in low-income and middle-income countries: an umbrella review
.
Lancet Psychiatry.
2020
;
7
(
2
):
162
172
.

61.

Fleischhacker
WWA.
Meta view on meta-analyses
.
JAMA Psychiatry.
2017
;
74
(
7
):
684
685
.

62.

Atkinson
AJ
,
Colburn
WA
,
DeGruttola
VG
, et al.
Biomarkers and surrogate endpoints: preferred definitions and conceptual framework
.
Clin Pharmacol Ther.
2001
;
69
(
3
):
89
95
.

63.

Ioannidis
JPA
,
Boffetta
P
,
Little
J
, et al.
Assessment of cumulative evidence on genetic associations: interim guidelines
.
Int J Epidemiol.
2008
;
37
:
120
132
.

64.

Picó-Pérez
M
,
Vieira
R
,
Fernández-rodríguez
M
,
De Barros
MAP
,
Radua
J
,
Morgado
P.
Multimodal meta-analysis of structural gray matter, neurocognitive and social cognitive fMRI findings in schizophrenia patients
.
Psychol Med.
2022
;
52
:
614
624
.

65.

Page
MJ
,
McKenzie
JE
,
Bossuyt
PM
, et al.
The PRISMA 2020 statement: an updated guideline for reporting systematic reviews
.
BMJ.
2021
;
372
(
71
):
n71
n79
.

66.

Shea
BJ
,
Grimshaw
JM
,
Wells
GA
, et al.
Development of AMSTAR: a measurement tool to assess the methodological quality of systematic reviews
.
BMC Med Res Methodol.
2007
;
7
:
10
.

67.

Gosling
CJ
,
Solanes
A
,
Fusar-Poli
P
,
Radua
J.
Metaumbrella: the first comprehensive suite to perform data analysis in umbrella reviews with stratification of the evidence
.
BMJ Ment Health.
2023
;
26
(
1
):
e300534
e300538
.

68.

Fullana
MA
,
Abramovitch
A
,
Via
E
, et al.
Diagnostic biomarkers for obsessive-compulsive disorder: a reasonable quest or ignis fatuus
?
Neurosci Biobehav Rev.
2020
;
118
:
504
513
.

69.

Solanes
A
,
Albajes-Eizagirre
A
,
Fullana
MA
, et al.
Can we increase the subjective well-being of the general population? An umbrella review of the evidence
.
Revista de psiquiatria y salud mental.
2021
;
14
(
1
):
50
64
.

70.

Fullana
MA
,
Tortella-Feliu
M
,
Fernández De La Cruz
L
, et al.
Risk and protective factors for anxiety and obsessive-compulsive disorders: an umbrella review of systematic reviews and meta-analyses
.
Psychol Med.
2020
;
50
(
8
):
1300
1315
.

71.

Hedges
LV.
Effect sizes in cluster-randomized designs
.
J Educ Behav Stat.
2007
;
32
(
4
):
341
370
.

72.

Egger
M
,
Smith
G
,
Schneider
M
,
Minder
C.
Bias in meta-analysis detected by a simple, graphical test
.
BMJ.
1997
;
315
(
7109
):
629
634
.

73.

Ioannidis
JPA.
Clarifications on the application and interpretation of the test for excess significance and its extensions
.
J Math Psychol.
2013
;
57
(
5
):
184
187
.

74.

Ezeoke
A
,
Mellor
A
,
Buckley
A
,
Miller
B.
A systematic, quantitative review of blood autoantibodies in schizophrenia
.
Schizophr Bull.
2013
;
150
(
1
):
245
251
.

75.

Wang
D
,
Zhai
J
,
Liu
D.
Serum folate levels in schizophrenia: a meta-analysis
.
Psychiatry Res.
2016
;
235
:
83
89
.

76.

Flatow
J
,
Buckley
P
,
Miller
BJ.
Meta-analysis of oxidative stress in schizophrenia
.
Biol Psychiatry.
2013
;
74
(
6
):
400
409
.

77.

Davison
J
,
Gorman
AO
,
Brennan
L
,
Cotter
DR.
A systematic review of metabolite biomarkers of schizophrenia
.
Schizophr Res.
2018
;
195
:
32
50
.

78.

Grignon
S
,
Marc
J.
Assessment of malondialdehyde levels in schizophrenia: a meta-analysis and some methodological considerations
.
Prog Neuropsychopharmacol Biol Psychiatry.
2007
;
31
:
365
369
.

79.

Cui
H
,
Jin
Y
,
Wang
J
,
Weng
X
,
Li
C.
Serum brain-derived neurotrophic factor (BDNF) levels in schizophrenia: a systematic review
.
Shanghai Arch Psychiatry.
2012
;
24
(
5
):
250
261
.

80.

Green
MJ
,
Matheson
SL
,
Shepherd
A
,
Weickert
CS
,
Carr
VJ.
Brain-derived neurotrophic factor levels in schizophrenia: a systematic review with meta-analysis
.
Mol Psychiatry.
2011
;
16
:
960
972
.

81.

Rowbotham
IM
,
Orsucci
FF
,
Mansour
MF
,
Chamberlain
SR
,
Raja
HY.
Relevance of brain-derived neurotrophic factor levels in schizophrenia: a systematic review and meta-analysis
.
AIMS Neurosci.
2015
;
2
(
4
):
280
293
.

82.

Toll
A
,
Mané
A.
Brain-derived neurotrophic factor levels in first episode of psychosis: a systematic review
.
World J Psychiatry.
2015
;
5
(
1
):
154
159
.

83.

Nishi
A
,
Numata
S
,
Tajima
A
, et al.
Meta-analyses of blood homocysteine levels for gender and genetic association studies of the MTHFR C677T polymorphism in schizophrenia
.
Schizophr Bull.
2014
;
40
(
5
):
1154
1163
.

84.

Haigh
SM
,
Coffman
BA
,
Salisbury
DF.
Mismatch negativity in first-episode schizophrenia: a meta-analysis
.
Clin EEG Neurosci.
2017
;
48
(
1
):
3
10
.

85.

Avissar
M
,
Xie
S
,
Vail
B
,
Lopez-Calderon
J
,
Wang
Y
,
Javitt
DC.
Meta-analysis of mismatch negativity to simple versus complex deviants in schizophrenia
.
Schizophr Res.
2018
;
191
:
25
34
.

86.

Umbricht
D
,
Krljes
S.
Mismatch negativity in schizophrenia: a meta-analysis
.
Schizophr Res.
2005
;
76
:
1
23
.

87.

Xiong
Y
,
Li
X
,
Zhao
L
,
Wang
C.
Mismatch negativity in Han Chinese patients with schizophrenia: a meta-analysis
.
Shanghai Arch Psychiatry.
2017
;
29
(
5
):
259
267
.

88.

Bramon
E
,
Rabe-Hesketh
S
,
Sham
P
,
Murray
RM
,
Frangou
S.
Meta-analysis of the P300 and P50 waveforms in schizophrenia
.
Schizophr Res.
2004
;
70
:
315
329
.

89.

Qiu
Y
,
Tang
Y
,
Chan
RCK
,
Sun
X
,
He
J.
P300 aberration in first-episode schizophrenia patients: a meta-analysis
.
PLoS One.
2014
;
9
(
6
):
e97794
e97798
.

90.

Buoli
M
,
Caldiroli
A
,
Melter
CC
,
Serati
M
,
de Nijs
J
,
Altamura
AC.
Biological aspects and candidate biomarkers for psychotic bipolar disorder: a systematic review
.
Psychiatry Clin Neurosci.
2016
;
70
:
227
244
.

91.

Cheng
C
,
Chan
P-YS
,
Liu
C
,
Hsu
S.
Auditory sensory gating in patients with bipolar disorders: a meta-analysis
.
J Affect Disord.
2016
;
203
:
199
203
.

92.

de Wilde
OM
,
Bour
LJ
,
Dingemans
PM
,
Koelman
JHTM
,
Linszen
DH.
A meta-analysis of P50 studies in patients with schizophrenia and relatives: differences in methodology between research groups
.
Schizophr Res.
2007
;
97
(
1–3
):
137
151
.

93.

Patterson
JV
,
Hetrick
WP
,
Boutros
NN
, et al.
P50 sensory gating ratios in schizophrenics and controls: a review and data analysis
.
Psychiatry Res.
2008
;
158
:
226
247
.

94.

Sayo
A
,
Jennings
RG
,
van Horn
JD.
Study factors influencing ventricular enlargement in schizophrenia: a 20 year follow-up meta-analysis
.
Neuroimage.
2012
;
59
(
1
):
154
167
.

95.

Brugger
S
,
Davis
JM
,
Leucht
S
,
Stone
JM.
Proton magnetic resonance spectroscopy and illness stage in schizophrenia—a systematic review and meta-analysis
.
Biol Psychiatry.
2011
;
69
(
5
):
495
503
.

96.

Xu
T
,
Chan
RCK
,
Compton
MT.
Minor physical anomalies in patients with schizophrenia, unaffected first-degree relatives, and healthy controls: a meta-analysis
.
PLoS One.
2011
;
6
(
9
):
e24129
e24127
.

97.

Clamor
A
,
Lincoln
TM
,
Thayer
JF
,
Koenig
J.
Resting vagal activity in schizophrenia: meta-analysis of heart rate variability as a potential endophenotype
.
Br J Psychiatry.
2016
;
208
:
9
16
.

98.

Di Sabatino
A
,
Vanoli
A
,
Giuffrida
P
,
Luinetti
O
,
Solcia
E
,
Corazza
GR.
The function of tissue transglutaminase in celiac disease
.
Autoimmun Rev.
2012
;
11
(
10
):
746
753
.

99.

Wijarnpreecha
K
,
Jaruvongvanich
V
,
Cheungpasitporn
W
,
Ungprasert
P.
Association between celiac disease and schizophrenia: a meta-analysis
.
Eur J Gastroenterol Hepatol.
2018
;
30
:
442
446
.

100.

Bender
L.
Childhood schizophrenia
.
Psychiatr Q.
1953
;
27
:
663
681
.

101.

Dohan
FC.
Wheat “consumption” and hospital admissions for schizophrenia during World War II. A preliminary report
.
Am J Clin Nutr.
1966
;
18
:
7
10
.

102.

Casella
G
,
Bordo
BM
,
Schalling
R
, et al.
Neurological disorders and celiac disease
.
Minerva Gastroenterol Dietol.
2015
;
61
:
1
2
.

103.

Zingone
F
,
Swift
GL
,
Card
TR
,
Sanders
DS
,
Ludvigsson
JF
,
Bai
JC.
Psychological morbidity of celiac disease: a review of the literature
.
United European Gastroenterol J.
2015
;
3
(
2
):
136
145
.

104.

Al-diwani
AAJ
,
Pollak
A
,
Irani
SR
,
Lennox
BR.
Psychosis: an autoimmune disease
?
Immunology.
2017
;
152
:
388
401
.

105.

Pollak
TA
,
Lennox
BR
,
Müller
S
, et al.
Autoimmune psychosis: an international consensus on an approach to the diagnosis and management of psychosis of suspected autoimmune origin
.
Lancet Psychiatry.
2020
;
7
:
93
108
.

106.

Kalaydjian
AE
,
Eaton
W
,
Cascella
N
,
The
FA.
The gluten connection: the association between schizophrenia and celiac disease
.
Acta Psychiatr Scand.
2006
;
113
:
82
90
.

107.

Stevens
AJ
,
Rucklidge
JJ
,
Kennedy
MA.
Epigenetics, nutrition and mental health. Is there a relationship
?
Nutr Neurosci.
2018
;
21
(
9
):
602
613
.

108.

Bottiglieri
T.
Homocysteine and folate metabolism in depression
.
Prog Neuropsychopharmacol Biol Psychiatry.
2005
;
29
:
1103
1112
.

109.

Hsieh
Y
,
Chou
L
,
Lin
C
,
Wu
H
,
Li
D
,
tseng
PT.
Serum folate levels in bipolar disorder: a systematic review and meta-analysis
.
BMC Psychiatry.
2019
;
19
(
305
):
1
9
.

110.

Davies
C
,
Segre
G
,
Estradé
A
, et al.
Prenatal and perinatal risk and protective factors for psychosis: a systematic review and meta-analysis
.
Lancet Psychiatry.
2020
;
7
(
5
):
399
410
.

111.

Kirkbride
JB
,
Susser
E
,
Kundakovic
M
,
Kresovich
JK
,
Davey Smith
G
,
Relton
CL.
Prenatal nutrition, epigenetics and schizophrenia risk: can we test causal effects
?
Epigenomics.
2012
;
4
(
3
):
303
315
.

112.

Wolff
GL
,
Kodell
RL
,
Moore
SR
,
Cooney
CA.
Maternal epigenetics and methyl supplements affect agouti gene expression in Avy/a mice
.
FASEB J.
1998
;
12
(
11
):
949
957
.

113.

Heijmans
BT
,
Tobi
EW
,
Stein
AD
, et al.
Persistent epigenetic differences associated with prenatal exposure to famine in humans
.
Proc Natl Acad Sci USA.
2008
;
105
(
44
):
17046
17049
.

114.

Cao
B
,
Wang
DF
,
Xu
MY
, et al.
Vitamin B12 and the risk of schizophrenia: a meta-analysis
.
Schizophr Res.
2016
;
172
(
1–3
):
216
217
.

115.

Zhang
Y
,
Hodgson
NW
,
Trivedi
MS
, et al.
Decreased brain levels of vitamin B12 in aging, autism and schizophrenia
.
PLoS One.
2016
;
11
(
1
):
e0146797
e0146719
.

116.

Aucoin
M
,
Lachance
L
,
Cooley
K
,
Kidd
S.
Diet and psychosis: a scoping review
.
Neuropsychobiology.
2020
;
79
(
1
):
20
42
.

117.

Misiak
B
,
Frydecka
D
,
Łaczmański
L
,
Ślęzak
R
,
Kiejna
A.
Effects of second-generation antipsychotics on selected markers of one-carbon metabolism and metabolic syndrome components in first-episode schizophrenia patients
.
Eur J Clin Pharmacol.
2014
;
70
(
12
):
1433
1441
.

118.

Moustafa
AA
,
Hewedi
DH
,
Eissa
AM
,
Frydecka
D
,
Misiak
B.
Homocysteine levels in schizophrenia and affective disorders—focus on cognition
.
Front Behav Neurosci.
2014
;
8
:
1
10
.

119.

Brown
AS
,
Bottiglieri
T
,
Schaefer
CA
, et al.
Elevated prenatal homocysteine levels as a risk factor for schizophrenia
.
Arch Gen Psychiatry.
2007
;
64
(
1
):
31
39
.

120.

Bouaziz
N
,
Ayedi
I
,
Sidhom
O
, et al.
Plasma homocysteine in schizophrenia: determinants and clinical correlations in Tunisian patients free from antipsychotics
.
Psychiatry Res.
2010
;
179
(
1
):
24
29
.

121.

Petronijević
ND
,
Radonjić
NV
,
Ivković
MD
, et al.
Plasma homocysteine levels in young male patients in the exacerbation and remission phase of schizophrenia
.
Prog Neuro-psychopharmacol Biol Psychiatry.
2008
;
32
(
8
):
1921
1926
.

122.

Goff
DC
,
Bottiglieri
T
,
Arning
E
, et al.
Folate, homocysteine, and negative symptoms in schizophrenia
.
Am J Psychiatry.
2004
;
161
:
1705
1708
.

123.

Di Lorenzo
R
,
Amoretti
A
,
Baldini
S
, et al.
Homocysteine levels in schizophrenia patients newly admitted to an acute psychiatric ward
.
Acta Neuropsychiatr.
2015
;
27
(
6
):
336
344
.

124.

Applebaum
J
,
Shimon
H
,
Sela
BA
,
Belmaker
RH
,
Levine
J.
Homocysteine levels in newly admitted schizophrenic patients
.
J Psychiatr Res.
2004
;
38
(
4
):
413
416
.

125.

Fraguas
D
,
Díaz-caneja
CM
,
Ayora
M
, et al.
Oxidative stress and inflammation in first-episode psychosis: a systematic review and meta-analysis
.
Schizophr Bull.
2019
;
45
(
4
):
742
751
.

126.

Onozato
M
,
Uta
A
,
Magarida
A
, 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
:
48
53
.

127.

Kinoshita
M
,
Numata
S
,
Tajima
A
,
Shimodera
S
,
Imoto
I
,
Ohmori
T.
Plasma total homocysteine is associated with DNA methylation in patients with schizophrenia
.
Epigenetics.
2013
;
8
(
6
):
584
590
.

128.

Tyagi
N
,
Sedoris
KC
,
Steed
M
,
Ovechkin
AV
,
Moshal
KS
,
Tyagi
SC.
Mechanisms of homocysteine-induced oxidative stress
.
Am J Physiol Hear Circ Physiol.
2005
;
289
:
H2649
H2656
.

129.

Zheng
W
,
Li
W
,
Qi
H
, et al.
Adjunctive folate for major mental disorders: a systematic review
.
J Affect Disord.
2020
;
267
:
123
130
.

130.

Firth
J
,
Stubbs
B
,
Sarris
J
, et al.
The effects of vitamin and mineral supplementation on symptoms of schizophrenia: a systematic review and meta-analysis
.
Psychol Med.
2017
;
47
(
9
):
1515
1527
.

131.

Sakuma
K
,
Matsunaga
S
,
Nomura
I
,
Okuya
M
,
Kishi
T
,
Iwata
N.
Folic acid/methylfolate for the treatment of psychopathology in schizophrenia: a systematic review and meta-analysis
.
Psychopharmacology (Berl).
2018
;
235
:
2303
2314
.

132.

Allott
K
,
McGorry
PD
,
Yuen
HP
, et al.
The vitamins in psychosis study: a randomized, double-blind, placebo-controlled trial of the effects of vitamins B12, B6, and folic acid on symptoms and neurocognition in first-episode psychosis
.
Biol Psychiatry.
2019
;
86
(
1
):
35
44
.

133.

Johnson
WG.
DNA polymorphism-diet-cofactor-development hypothesis and the gene-teratogen model for schizophrenia and other developmental disorders
.
Am J Med Genet.
1999
;
88
:
311
323
.

134.

Hill
M
,
Shannahan
K
,
Jasinski
S
, et al.
Folate supplementation in schizophrenia: a possible role for MTHFR genotype
.
Schizophr Res.
2011
;
127
(
1–3
):
41
45
.

135.

Roffman
JL
,
Brohawn
DG
,
Nitenson
AZ
,
MacKlin
EA
,
Smoller
JW
,
Goff
DC.
Genetic variation throughout the folate metabolic pathway influences negative symptom severity in schizophrenia
.
Schizophr Bull.
2013
;
39
(
2
):
330
338
.

136.

Landek-salgado
MA
,
Faust
TE
,
Sawa
A.
Molecular substrates of schizophrenia: homeostatic signaling to connectivity
.
Mol Psychiatry.
2016
;
21
:
10
28
.

137.

Schizophrenia Working Group of the Psychiatric Genomics Consortium
.
Biological insights from 108 schizophrenia-associated genetic loci
.
Nature.
2014
;
24
(
511
):
421
427
.

138.

Pouget
JG
,
Gonçalves
VF
,
Spain
SL
, et al. ;
Schizophrenia Working Group of the Psychiatric Genomics Consortium
.
Genome-wide association studies suggest limited immune gene enrichment in schizophrenia compared to 5 autoimmune diseases
.
Schizophr Bull.
2016
;
42
(
5
):
1176
1184
.

139.

Trépanier
MO
,
Hopperton
KE
,
Mizrahi
R
,
Mechawar
N
,
Bazinet
RP.
Postmortem evidence of cerebral in flammation in schizophrenia: a systematic review
.
Mol Psychiatry.
2016
;
21
:
1009
1026
.

140.

Rodrigues-Amorim
D
,
Rivera-baltanás
T
,
Spuch
C
, et al.
Cytokines dysregulation in schizophrenia: a systematic review of psychoneuroimmune relationship
.
Schizophr Res.
2018
;
197
:
19
33
.

141.

Baumeister
D
,
Akhtar
R
,
Ciufolini
S
,
Pariante
CM
,
Mondelli
V.
Childhood trauma and adulthood in flammation: a meta-analysis of peripheral C-reactive protein, interleukin-6 and tumour necrosis factor-α
.
Mol Psychiatry.
2016
;
21
:
642
649
.

142.

Khoury
R
,
Nasrallah
HA.
Inflammatory biomarkers in individuals at clinical high risk for psychosis (CHR-P): state or trait
?
Schizophr Res.
2018
;
199
:
31
38
.

143.

Goldsmith
DR
,
Rapaport
MH
,
Miller
BJ.
A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression
.
Mol Psychiatry.
2016
;
21
(
12
):
1696
1709
.

144.

Çakici
N
,
van Beveren
NJM
,
Judge-Hundal
G
,
Koola
MM
,
Sommer
IE.
An update on the efficacy of anti-inflammatory agents for patients with schizophrenia: a meta-analysis
.
Psychol Med.
2019
;
49
:
2307
2319
.

145.

Martinez-Cengotitabengoa
M
,
MacDowell
K
,
Alberich
S
, et al. ;
FLAMM-PEPs
.
BDNF and NGF signalling in early phases of psychosis: relationship with inflammation and response to antipsychotics after 1 year
.
Schizophr Bull.
2016
;
42
(
1
):
sbv078
sbv151
.

146.

Erickson
MA
,
Ruf
A
,
Gold
JM.
A meta-analysis of mismatch negativity in schizophrenia: from clinical risk to disease specificity and progression
.
Biol Psychiatry.
2016
;
79
:
980
987
.

147.

Umbricht
D
,
Koller
R
,
Vollenweider
FX
,
Schmid
L.
Mismatch negativity predicts psychotic experiences induced by NMDA receptor antagonist in healthy volunteers
.
Biol Psychiatry.
2002
;
51
(
5
):
400
406
.

148.

Javitt
DC
,
Steinschneidertt
M
,
Schroedertt
CE
,
Arezzott
JC.
Role of cortical N-methyl-d-aspartate receptors in auditory sensory memory and mismatch negativity generation: implications for schizophrenia
.
Proc Natl Acad Sci.
1996
;
93
:
11962
11967
.

149.

Hay
RA
,
Roach
BJ
,
Srihari
VH
,
Woods
SW
,
Ford
JM
,
Mathalon
DH.
Equivalent mismatch negativity deficits across deviant types in early illness schizophrenia-spectrum patients
.
Biol Psychol.
2015
;
105
:
130
137
.

150.

Hirt
V
,
Schubring
D
,
Schalinski
I
,
Rockstroh
B.
Mismatch negativity and cognitive performance in the course of schizophrenia
.
Int J Psychophysiol.
2019
;
145
:
30
39
.

151.

Bodatsch
M
,
Ruhrmann
S
,
Wagner
M
, et al.
Prediction of psychosis by mismatch negativity
.
Biol Psychiatry.
2010
;
69
(
10
):
959
966
.

152.

Shaikh
M
,
Valmaggia
L
,
Broome
MR
, et al.
Reduced mismatch negativity predates the onset of psychosis
.
Schizophr Res.
2012
;
134
(
1
):
42
48
.

153.

Higuchi
Y
,
Sumiyoshi
T
,
Seo
T
,
Miyanishi
T
,
Kawasaki
Y
,
Suzuki
M.
Mismatch negativity and cognitive performance for the prediction of psychosis in subjects with at-risk mental state
.
PLoS One.
2013
;
8
(
1
):
e54010
e54080
.

154.

Solís-vivanco
R
,
Mondragón-maya
A
,
León-ortiz
P
,
Rodríguez-agudelo
Y
,
Cadenhead
KS
,
Fuente-sandoval
CD.
Mismatch Negativity reduction in the left cortical regions in first-episode psychosis and in individuals at ultra high-risk for psychosis
.
Schizophr Res.
2014
;
158
(
1–3
):
58
63
.

155.

Nagai
T
,
Tada
M
,
Kirihara
K
,
Araki
T
,
Jinde
S
,
Kasai
K.
Mismatch negativity as a “translatable” brain marker toward early intervention for psychosis: a review
.
Front Psychiatry.
2013
;
4
:
1
10
.

156.

McCarthy
G
,
Donchin
E.
A metric for thought: a comparison of P300 latency and reaction time
.
Science (New York, N.Y.).
1981
;
211
:
77
80
.

157.

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
.

158.

Polich
J
,
Kokb
A.
Cognitive and biological determinants of P300: an integrative review
.
Biol Psychol
.
1995
;
41
:
103
146
.

159.

Wang
J
,
Tang
Y
,
Li
C
, et al.
Decreased P300 current source density in drug-naive first episode schizophrenics revealed by high density recording
.
Int J Psychophysiol.
2010
;
75
:
249
257
.

160.

del Re
EC
,
Spencer
KM
,
Oribe
N
, et al.
Clinical high risk and first episode schizophrenia: auditory event-related potentials
.
Psychiatry Res.
2015
;
231
(
2
):
126
133
.

161.

McCarley
RW
,
Salisbury
DF
,
Hirayasu
Y
, et al.
Association between smaller left posterior superior temporal gyrus volume on magnetic resonance imaging and smaller left temporal P300 amplitude in first-episode schizophrenia
.
Arch Gen Psychiatry.
2002
;
59
:
321
331
.

162.

Jeon
YW
,
Polich
J.
Meta-analysis of P300 and schizophrenia: patients, paradigms, and practical implications
.
Psychophysiology.
2003
;
40
:
684
701
.

163.

Kim
M
,
Lee
TH
,
Kim
JH
, 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
.

164.

Park
E
,
Han
S
,
Jeon
Y.
Auditory and visual P300 reflecting cognitive improvement in patients with schizophrenia with quetiapine: a pilot study
.
Prog Neuropsychopharmacol Biol Psychiatry.
2010
;
34
:
674
680
.

165.

Fusar-poli
P
,
Crossley
N
,
Woolley
J
, et al.
Gray matter alterations related to P300 abnormalities in subjects at high risk for psychosis: longitudinal MRI-EEG study
.
Neuroimage.
2011
;
55
(
1
):
320
328
.

166.

Fusar-poli
P
,
Crossley
N
,
Woolley
J
, et al.
White matter alterations related to P300 abnormalities in individuals at high risk for psychosis: an MRI—EEG study
.
J Psychiatry Neurosci.
2011
;
36
(
4
):
239
248
.

167.

Tang
Y
,
Wang
J
,
Zhang
T
, et al.
P300 as an index of transition to psychosis and of remission: data from a clinical high risk for psychosis study and review of literature
.
Schizophr Res.
2020
;
226
:
74
83
.

168.

Hamilton
HK
,
Roach
BJ
,
Bachman
PM
, et al.
Association between P300 responses to auditory oddball stimuli and clinical outcomes in the psychosis risk syndrome
.
JAMA Psychiatry.
2019
;
76
(
11
):
1187
1197
.

169.

Bodatsch
M
,
Brockhaus-Dumke
A
,
Klosterkötter
J
,
Ruhrmann
S.
Forecasting psychosis by event-related potentials—systematic review and specific meta-analysis
.
Biol Psychiatry.
2015
;
77
(
11
):
951
958
.

170.

Catalan
A
,
Salazar de Pablo
G
,
Aymerich
C
, et al.
Neurocognitive functioning in individuals at clinical high risk for psychosis: systematic review and meta-analysis
.
JAMA Psychiatry.
2021
;
78
:
859
.

171.

Fortea
A
,
Batalla
A
,
Radua
J
, et al.
Cortical gray matter reduction precedes transition to psychosis in individuals at clinical high-risk for psychosis: a voxel-based meta-analysis
.
Schizophr Res.
2021
;
232
:
98
106
.

172.

Johnstone
E
,
Frith
CD
,
Crow
TJ
,
Husband
J
,
Kreel
L.
Cerebral ventricular size and cognitive impairment in chronic schizophrenia
.
Lancet.
1976
;
308
(
7992
):
924
926
.

173.

Reveley
A
,
Clifford
CA
,
Reveley
MA
,
Murray
RM.
Cerebral ventricular size in twins discordant for schizophrenia
.
Lancet.
1982
;
319
(
8271
):
540
541
.

174.

Pfefferbaum
A
,
Zipursky
RB
,
Lim
KO
,
Zatz
LM
,
Stahl
SM
,
Jernigan
TL.
Computed tomographic evidence for generalized sulcal and ventricular enlargement in schizophrenia
.
Arch Gen Psychiatry.
1988
;
45
:
633
640
.

175.

Weinberger
DR
,
Fuller Torrey
E
,
Neophytides
AN
,
Wyatt
RJ.
Structural abnormalities in the cerebral cortex of chronic schizophrenic patients
.
Arch Gen Psychiatry.
1979
;
36
:
935
939
.

176.

McDonald
C
,
Marshall
N
,
Sham
PC
, et al.
Regional brain morphometry in patients with unaffected relatives
.
Am J Psychiatry.
2006
;
163
:
478
487
.

177.

Chung
Y
,
Haut
KM
,
He
G
, et al. ;
North American Prodrome Longitudinal Study (NAPLS) Consortium
.
Ventricular enlargement and progressive reduction of cortical gray matter are linked in prodromal youth who develop psychosis
.
Schizophr Res.
2017
;
189
:
169
174
.

178.

Mondino
M
,
Brunelin
J
,
Saoud
M.
N-acetyl-aspartate level is decreased in the prefrontal cortex in subjects at-risk for schizophrenia
.
Front Psychiatry.
2013
;
4
:
1
6
.

179.

Natsubori
T
,
Inoue
H
,
Abe
O
, et al.
Reduced frontal glutamate + glutamine and N-acetylaspartate levels in patients with chronic schizophrenia but not in those at clinical high risk for psychosis or with first-episode schizophrenia
.
Schizophr Bull.
2014
;
40
(
5
):
1128
1139
.

180.

Smieskova
R
,
Fusar-Poli
P
,
Allen
P
, et al.
Neuroscience and biobehavioral reviews neuroimaging predictors of transition to psychosis—a systematic review and meta-analysis
.
Neurosci Biobehav Rev.
2010
;
34
(
8
):
1207
1222
.

181.

Mechelli
A
,
Riecher-Rössler
A
,
Meisenzahl
EM
, et al.
Neuroanatomical abnormalities that predate the onset of psychosis
.
Arch Gen Psychiatry.
2011
;
68
(
5
):
489
495
.

182.

Fusar-Poli
P
,
Salazar De Pablo
G
,
Correll
CU
, et al.
Prevention of psychosis: advances in detection, prognosis, and intervention
.
JAMA Psychiatry.
2020
;
77
(
7
):
755
765
.

183.

Zipursky
RB
,
Reilly
TJ
,
Murray
RM.
The myth of schizophrenia as a progressive brain disease
.
Schizophr Bull.
2013
;
39
(
6
):
1363
1372
.

184.

Fusar-Poli
P
,
Smieskova
R
,
Kempton
MJ
,
Ho
BC
,
Andreasen
NC
,
Borgwardt
S.
Progressive brain changes in schizophrenia related to antipsychotic treatment? A meta-analysis of longitudinal MRI studies
.
Neurosci Biobehav Rev.
2013
;
37
(
8
):
1680
1691
.

185.

Sayo
A
,
Jennings
RG
,
Van Horn
JD.
Study factors in fluencing ventricular enlargement in schizophrenia: a 20 year follow-up meta-analysis
.
Neuroimage.
2012
;
59
:
154
167
.

186.

Walther
S
,
Strik
W.
Motor symptoms and schizophrenia
.
Neuropsychobiology.
2012
;
66
:
77
92
.

187.

Reichenberg
A
,
Caspi
A
,
Harrington
H
, et al.
Static and dynamic cognitive deficits in childhood preceding adult schizophrenia: a 30-year study
.
Am J Psychiatry.
2010
;
167
:
160
169
.

188.

Fusar-poli
P
,
Borgwardt
S
,
Crescini
A
, et al.
Neuroanatomy of vulnerability to psychosis: a voxel-based meta-analysis
.
Neurosci Biobehav Rev.
2011
;
35
:
1175
1185
.

189.

Koutsouleris
N
,
Riecher-Rössler
A
,
Meisenzahl
EM
, et al.
Detecting the psychosis prodrome across high-risk populations using neuroanatomical biomarkers
.
Schizophr Bull.
2015
;
41
(
2
):
471
482
.

190.

Murray
RM
,
Lewis
SW.
Is schizophrenia a neurodevelopmental disorder
?
J R Army Med Corps.
1987
;
295
(
3
):
681
682
.

191.

Weinberger
DR.
Implications of normal brain development for the pathogenesis of schizphrenia
.
Arch Gen Psychiatry.
1987
;
44
(
7
):
660
669
.

192.

Murray
RM
,
Bhavsar
V
,
Tripoli
G
,
Howes
O.
30 years on: how the neurodevelopmental hypothesis of schizophrenia morphed into the developmental risk factor model of psychosis
.
Schizophr Bull.
2017
;
43
(
6
):
1190
1196
.

193.

Lill
A
,
Helge
B
,
Thayer
JF.
Vagal influence on working memory and attention
.
Int J Psychophysiol.
2003
;
48
:
263
274
.

194.

Thayer
JF
,
Lane
RD.
A model of neurovisceral integration in emotion regulation and dysregulation
.
J Affect Disord.
2000
;
61
:
201
216
.

195.

Williams
DP
,
Cash
C
,
Rankin
C
,
Bernardi
A
,
Thayer
JF.
Resting heart rate variability predicts self-reported difficulties in emotion regulation: a focus on different facets of emotion regulation
.
Front Psychol.
2015
;
6
:
1
8
.

196.

Thayer
JF
,
Hansen
AL
,
Saus-Rose
E
,
Johnsen
BH.
Heart rate variability, prefrontal neural function, and cognitive performance: the neurovisceral integration perspective on self-regulation, adaptation, and health
.
Ann Behav Med.
2009
;
37
:
141
153
.

197.

Hennekens
CH
,
Hennekens
AR
,
Hollar
D
,
Casey
DE.
Schizophrenia and increased risks of cardiovascular disease
.
Am Heart J.
2005
;
150
:
1115
1121
.

198.

Clamor
A
,
Sundag
J
,
Lincoln
TM.
Specificity of resting-state heart rate variability in psychosis: a comparison with clinical high risk, anxiety, and healthy controls
.
Schizophr Res.
2019
;
206
:
89
95
.

199.

Kocsis
A
,
Gajwani
R
,
Gross
J
, et al.
Altered autonomic function in individuals at clinical high risk for psychosis
.
Front Psychiatry.
2020
;
11
:
1
11
.

200.

Van Os
J
,
Kenis
G
,
Rutten
BPF.
The environment and schizophrenia
.
Nature.
2010
;
468
(
7321
):
203
212
.

201.

Feczko
E
,
Miranda-Dominguez
O
,
Marr
M
,
Graham
AM
,
Nigg
JT
,
Fair
DA
The heterogeneity problem: approaches to identify psychiatric subtypes
.
Trends Cogn Sci.
2019
;
23
(
7
):
584
601
.

202.

Ioannidis
JPA.
Why most discovered true associations are inflated
.
Epidemiology.
2008
;
19
(
5
):
640
648
.

203.

Carvalho
AF
,
Köhler
CA
,
Brunoni
AR
, et al.
Bias in peripheral depression biomarkers
.
Psychother Psychosom.
2016
;
85
(
2
):
81
90
.

204.

Carvalho
AF
,
Köhler
CA
,
Fernandes
BS
, et al.
Bias in emerging biomarkers for bipolar disorder
.
Psychol Med.
2016
;
46
(
11
):
2287
2297
.

205.

Oliver
D.
The importance of external validation to advance precision psychiatry
.
Lancet Reg Health Eur.
2022
;
22
:
100498
.

206.

Fusar-poli
P
,
Manchia
M
,
Koutsouleris
N
, et al. ;
PSMD EBRA cluster (annex 1)
.
Ethical considerations for precision psychiatry: a roadmap for research and clinical practice
.
Eur Neuropsychopharmacol.
2022
;
63
:
17
34
.

207.

Kéri
P
,
White
LA
,
Oliver
D
,
Fusar-Poli
P.
Empowering experts by experience to guide ethical precision psychiatry
.
Biol Psychiatry.
2023
;Preprint:
S0006-3223(23)01744
. doi:10.1016/j.biopsych.2023.11.015

208.

Addington
J
,
Liu
L
,
Brummitt
K
, et al.
North American Prodrome Longitudinal Study (NAPLS 3): methods and baseline description
.
Schizophr Res.
2022
;
243
:
262
267
.

209.

Brady
LS
,
Larrauri
CA
;
AMP SCZ Steering Committee
.
Accelerating Medicines Partnership® Schizophrenia (AMP®SCZ): developing tools to enable early intervention in the psychosis high risk state
.
World Psychiatry.
2023
;
22
(
1
):
42
43
.

210.

Andreazza
AC
,
Laksono
I
,
Fernandes
BS
, et al.
Guidelines for the standardized collection of blood-based biomarkers in psychiatry: steps for laboratory validity—a consensus of the biomarkers task force from the WFSBP
.
World J Biol Psychiatry.
2019
;
20
(
5
):
340
351
.

211.

Pisanu
C
,
Severino
G
,
De Toma
I
, et al. ;
European College of Neuropsychopharmacology (ECNP) Pharmacogenomics & Transcriptomics Network
.
Transcriptional biomarkers of response to pharmacological treatments in severe mental disorders: a systematic review
.
Eur Neuropsychopharmacol.
2022
;
55
:
112
157
.

Author notes

Paola Fuentes-Claramonte and Andrés Estradé Shared first authorship.

Joaquim Radua and Paolo Fusar-Poli Shared senior authorship.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.