Skip to main content

REVIEW article

Front. Aging Neurosci., 12 May 2023
Sec. Neuroinflammation and Neuropathy
Volume 15 - 2023 | https://doi.org/10.3389/fnagi.2023.1174644

Biomarkers of delirium risk in older adults: a systematic review and meta-analysis

  • 1Geriatric Unit, Hospital Universitario de Navarra (HUN), Pamplona, Spain
  • 2Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
  • 3Proteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
  • 4Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
  • 5CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
  • 6Orthopaedic Surgery and Traumatology Department, Clínica Universidad de Navarra, Pamplona, Spain

Delirium is a neuropsychiatric syndrome associated with increased morbidity and mortality in older patients. The aim of this study was to review predictive biomarkers of delirium in older patients to gain insights into the pathophysiology of this syndrome and provide guidance for future studies. Two authors independently and systematically searched MEDLINE, Embase, Cochrane Library, Web of Science and Scopus databases up to August 2021. A total of 32 studies were included. Only 6 studies were eligible for the meta-analysis, pooled results showed a significant increase in some serum biomarkers (C-reactive protein [CRP], tumour necrosis factor alpha [TNF-α] and interleukin-6 [IL-6]) among patients with delirium (odds ratio = 1.88, 95% CI 1.01 to 1.637; I2 = 76.75%). Although current evidence does not favour the use of any particular biomarker, serum CRP, TNF-α, and IL-6 were the most consistent biomarkers of delirium in older patients.

Introduction

Delirium is a severe neuropsychiatric syndrome characterized by an acute change in attention and other aspects of cognition such as altered arousal, disorientation, psychosis or mood disturbance (Sachdev et al., 2014). Approximately 23% of hospitalized older adults develop delirium in medical settings (Gibb et al., 2020); this proportion increases to 13–50% in noncardiac surgical settings (Inouye et al., 2014), 6–74% in inpatient palliative care units (Watt et al., 2019) and 50–70% among mechanically ventilated patients (Krewulak et al., 2018). Delirium is associated with multiple adverse outcomes, such as higher length of stay, higher risk of iatrogenic and medical complications during hospitalization (falls, malnutrition, dehydration, aspiration pneumonia, immobility, bedsores), increased disability, cognitive impairment, mental illnesses (depression, anxiety, posttraumatic stress disorder) and mortality (Abelha et al., 2013; Martínez-Velilla et al., 2016; Dharmarajan et al., 2017; Altman et al., 2018; Israni et al., 2018; Sillner et al., 2019; Welch et al., 2019; Hayhurst et al., 2020).

The risk of delirium is determined by predisposing factors (for example, pre-existing cognitive impairment, advanced age or frailty) and precipitating factors (acute insults such as surgery, infections or metabolic decompensations) (Wilson et al., 2020). Because multiple factors are implicated in the aetiology of delirium, there are likely several neurobiological processes that contribute to its pathophysiology, that remains unclear. During delirium, a reversal of the relationship between the dorsolateral prefrontal cortex (part of the executive network) (Choi et al., 2012) and the posterior cingulate cortex (involved in the default mode network) (Raichle, 2015) has been observed, contributing to changes in behaviour and attention. In addition, reduced brain efficiency and connectivity strength (especially in subcortical regions related to arousal) have been found in patients with delirium. Delirium can be defined as a failure of a vulnerable brain to show resilience in response to an acute stressor (van Montfort et al., 2019). This vulnerability can be caused by several processes such as impaired brain network connectivity which leads to neurotransmitter disturbance in cholinergic and noradrenergic neurons (Morandi et al., 2012), neuroinflammatory and glial cell changes which causes an exacerbated pro-inflammatory response to noxious insults (Murray et al., 2012) and vasculature dysfunction which produces endothelial injury, blood–brain barrier (BBB) damage and impaired brain perfusion (Cavallari et al., 2016).

Given the complexity of these processes, the use of biomarkers has become widespread for identification of delirium and its risk. The World Health Organization (WHO) defines a biomarker as any substance, structure, or process that can be measured in the body or its products that can influence or predict the incidence of outcome or disease [World Health Organization (WHO), 2001]. Biomarkers are objective and quantifiable characteristics of biological processes, which makes them a reliable and reproducible measure (Strimbu and Tavel, 2010). They are categorized into three patterns: (1) risk markers, which indicate the risk of a particular disease, (2) disease markers, which are correlated with the onset or recovery of a disease, and (3) end-products, which indicate disease resolution (Aronson and Ferner, 2017). The identification of biomarkers associated with delirium may help clarify its pathophysiology and aid in the prediction, diagnosis and management of this syndrome. Although few systematic reviews have been recently carried out on delirium biomarkers, they present important limitations, such as restriction to a single fluid (Hall et al., 2011, 2018; Ida et al., 2020) or a single biomarker (Liu et al., 2018; Narayanan et al., 2021), different types of mixed biomarkers (Chu et al., 2011; Dunne et al., 2021) and the absence of a focus on older adults, in whom delirium risk is the greatest.

Thus, the aim of this study was to systematically review the current literature on delirium biomarkers and identify risk markers in older adults to summarize the existing knowledge, enhance our understanding regarding the pathophysiology and provide methodological guidance for future studies.

Materials and methods

Search strategy

This systematic review was undertaken in accordance with the guidelines described by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) (Liberati et al., 2009). We asked the question whether incident delirium was associated with biomarkers and summarized data extracted from included studies.

Two reviewers (LL and BAC) searched studies published before 17 August 2021 that investigated delirium biomarkers. Searches were performed using a comprehensive text-word and Medical Subject Headings-based electronic search of MEDLINE, EMBASE, The Cochrane Library, Web of Science and Scopus. Primary key words included: “delirium” and “biomarker” used in combination with additional key words as: “cognitive dysfunction,” “acute confusion state,” “acute brain failure,” “postoperative delirium” or “postoperative cognitive disorder.” Boolean operators were used to combine search terms above (Supplementary material). Authors with specialist knowledge of the subject extracted the data. To avoid bias, results were based on analysis by LL, AC, NM, and AG. Variations in interpretation were reviewed, and any disputes were resolved by NM. The review protocol was registered in PROSPERO (CRD42021281272).

Selection criteria

The inclusion criteria were:

– Study design: case–control, cohort study or case series with non-delirious subjects as controls

– Language of publication: English or Spanish

– Year of publication: all studies until 17.08.2021

– Study subjects: mean patient aged ≥65 years

– Diagnostic criteria of delirium:

ο Diagnostic Statistical Manual of Mental Disorders (DSM)

ο International Classification of Diseases (ICD)

ο Delirium assessment tool based on DSM or ICD

– Source of biomarker: cerebrospinal fluid, blood or other body fluids.

– Measurement methods: methods that provide quantitative (i.e., ELISA, RIA) or detailed qualitative data (i.e., proteomics)

– Full text available (detailed information)

The exclusion criteria were:

– Reviews, case reports or comments, letters, personal opinions, book chapters and conference abstracts

– Randomized controlled trials (RCTs) measuring the effects of drugs on delirium incidence

– Studies where biomarkers were not identified

– Association between biomarkers and delirium in experimental studies (in vitro or in vivo animal studies)

– No identifiable delirium/no delirium subgroups

– Delirium of other specific causes (delirium tremens or other alcohol withdrawal states, Wernicke’s encephalopathy, neuropsychiatric systematic lupus erythematosus).

Data extraction and synthesis

Two authors independently screened the remaining literature for useable data and extracted it, and this was checked by another reviewer. Articles that met the inclusion criteria were reviewed and we recorded (if available) the following information: (1) Author and year of publication, (2) study design and setting (i.e., orthopedics, cardiac surgery, ICU patients…), (3) number and characteristics of patients included (age, sex, delirious/no delirious), (4) method used to diagnose delirium and delirium severity, (5) presence of cognitive impairment and method of assessment, (6) type of biomarker analyzed and levels, (7) method of obtaining the samples, (8) analytical laboratory methodology, and (9) main study findings. In cases in which more than one publication of the same trial existed, only the latest publication with the most complete data was included. Disagreements were settled by consensus.

In the case of studies that included both people younger and older than 65 years and specific data for older adults were included in the publication, we only used data from individual participants ≥65 years. If individual participant data for the subgroup of interest could not be obtained, we included studies if greater than or equal to 80% of the participants were ≥ 65 years.

Endpoints

The main risk markers of delirium are shown in Table 1.

TABLE 1
www.frontiersin.org

Table 1. Risk markers of delirium.

Assessment of study quality

Study quality and risk of bias were assessed independently by two of the authors using the Newcastle-Ottawa scale (NOS), which is designed for assessing common causes of bias in cohort studies. The NOS score ranges from 0 to 9 stars. A quality score was calculated based on three major components: (1) the selection of study groups (0–4 stars), (2) the comparability of study groups (0–2 stars), and (3) ascertainment of the exposure and outcome of interest. Overall, the quality of studies was deemed as poor (0 to 3 stars), fair (4 to 6 stars) or excellent (7 to 9 stars). Disagreement was resolved by discussion and consensus. The summary of assessment of risk of bias and additional comments on study quality are shown in Supplementary material.

Statistical analyses

All analyses were conducted using the DerSimonian-Laird random-effects inverse variance model using STATA software (version 17; StataCorp, College Station, TX, United States). Data were pooled only if biomarkers were reported in at least two studies. We used the odds ratios (OR) as the main effect size for the present study. We converted other estimations (e.g., standardized regression coefficients, standardized mean differences) to OR according to their corresponding formulas. A subgroup analysis according to inflammatory parameters was also included for CRP, TNF-α, and IL-6 data. Heterogeneity across studies was calculated using the inconsistency index (I2) (Cooper et al., 1994), and Egger’s regression intercept test was used to detect small-study effects bias (Egger et al., 1997; Higgins et al., 2003). No other sub-group analysis was performed due to the limited number of studies.

Results

Results of the literature search

We located 2,518 records, of which 1,644 remained after removing duplicates. After screening the titles and abstracts, 217 studies remained for full text review. Ultimately, 32 articles were included (shown in Figure 1). The major characteristics of the included studies are presented in Supplementary material, and a summary of the main results is shown in Table 2.

FIGURE 1
www.frontiersin.org

Figure 1. Flowchart.

TABLE 2
www.frontiersin.org

Table 2. Summary of main results of risk markers of delirium.

Study population

There was diversity between the clinical settings of the studies included in this systematic review. Most of them (30 studies) were carried out in surgical patients: 10 studies in cardiac surgery patients, 14 in orthopaedic surgery patients, 2 in cancer surgery and 4 in other surgery patients. Only 2 studies were conducted among medical inpatients (1 in the hospital ward and the other in the ICU).

The age of the patients included in the studies varied between 67 and 88 years. Seventeen studies included more males than females, while females were more prevalent in 15 studies.

Delirium assessment, delirium severity and delirium subtype

The Confusion Assessment Method was the most commonly used tool for assessing delirium (Inouye et al., 1990; Helfand et al., 2021), followed by the Confusion Assessment Method for Intensive Care Unit (CAM-ICU) (Gusmao-Flores et al., 2012), DSM and Delirium Rating Scale (DRS) (Trzepacz, 1999). Eighteen studies applied the CAM to define delirium, nine studies used the CAM-ICU, three studies assessed delirium with the DSM criteria, one study used the DRS and DSM criteria and one study used both the CAM and the CAM-ICU.

The severity of delirium was assessed in ten studies. The most commonly used tool was the Memorial Delirium Assessment Scale (MDAS), followed by the Confusion Assessment Method-Severity (CAM-S) and the DRS.

The subtype of delirium was only evaluated in one study, which used the Richmond Agitation-Sedation Scale (RASS).

Sample characteristics

Most of the studies collected blood samples. Cerebrospinal fluid (CSF) was collected in 4 studies and both blood and CSF were collected in 5 studies.

Risk markers of delirium

Neurotransmitters

Serum anticholinergic activity (SAA) was investigated as a risk marker of delirium in three studies. There were mixed results, as 2 studies (Cerejeira et al., 2012; Adam et al., 2020) demonstrated that lower preoperative acetylcholinesterase (AChE) activity was related to postoperative delirium (POD). One study showed that lower preoperative butyrylcholinesterase (BChE) activity was correlated with POD (Cerejeira et al., 2012), but another study did not find an association (Adam et al., 2020). Low levels of acetylcholine (Ach) before surgery were an independent risk factor for POD (Ma et al., 2020). de Jonghe et al. (2012) found that a higher preoperative kynurenine/tryptophan ratio and higher indoleamine 2,3-dioxygenase (IDO) activity were risk markers for POD. Higher postoperative levels of homovanillic acid (HVA) were associated with POD (Osse et al., 2012).

Hormones

Three papers explored a link between serum hormones and incident delirium. One study found that medical inpatients with higher levels of estradiol had an increased risk of developing delirium (Avila-Funes et al., 2015). Two studies investigated cortisol; Ma et al. (2020) found that higher preoperative cortisol levels were an independent risk factor for delirium, but Avila-Funes et al. (2015) did not find any associations between cortisol in medical inpatients and incident delirium. Li et al. (2017) found that low leptin levels at ICU admission were independently associated with delirium.

Neuronal damage biomarkers

Five studies focused on the relationship between biomarkers of neuronal damage and delirium. Hov et al. (2017) found that higher preoperative levels of S100 calcium-binding protein B (S100B) in CSF correlated with POD in patients who also had pathological levels of p-tau. Serum Neurofilament light (NfL) was assessed in two studies. Fong et al. (2020) found that higher preoperative levels were associated with POD and POD severity, but Saller et al. (2019) did not find differences. Szwed et al. (2020) evaluated serum phosphorylated axonal neurofilament subunit H (pNfLH), but no association with delirium was found. One study investigated serum ubiquitin carboxyl-terminal hydrolase L1 (UCHL-1) (Fong et al., 2020) and another study examined CSF neurogranin (Halaas et al., 2021), but no differences were reported between these biomarkers and delirium.

Neuroinflammatory markers

Nineteen studies analysed the role of biomarkers of neuroinflammation in delirium. The most investigated biomarkers in this field were C-reactive protein (CRP), tumour necrosis factor (TNF) and interleukin-6 (IL-6). Seven studies explored CRP. Five studies found that higher preoperative CRP in serum correlated with POD (Cerejeira et al., 2012; Shen et al., 2016; Dillon et al., 2017; Vasunilashorn et al., 2019; Chen et al., 2020); however, two studies did not find differences (Çinar et al., 2014; Katsumi et al., 2020). Kaźmierski et al. (2021) found that high levels of high-sensitivity C-reactive protein (hsCRP) in serum were also associated with POD, and Peng et al. (2019) also analysed the C-reactive protein-to-albumin ratio (CAR), showing that higher preoperative levels of serum CAR were an independent risk factor for POD. TNF-α was analysed in six studies; only one found that higher preoperative levels of TNF-α were related to POD but not significantly (Peng et al., 2019); in the rest of the studies, no differences were found (Cerejeira et al., 2012; Çinar et al., 2014; Hirsch et al., 2016; Chen et al., 2020; Sajjad et al., 2020). Seven studies investigated IL-6: four studies found that higher preoperative levels of IL-6 were related to delirium (Hirsch et al., 2016; Shen et al., 2016; Chen et al., 2019; Peng et al., 2019), while three studies found no differences (Cerejeira et al., 2012; Chen et al., 2020; Katsumi et al., 2020).

Other interleukins were explored, such as IL-1ra (Cape et al., 2014), IL-2 (Hirsch et al., 2016) and IL-10 (Hirsch et al., 2016), but they were not found to be related to the incidence of delirium. Cape et al. (2014) found that higher IL-1ß in CSF correlated with POD, but in two other studies, no association was found (Cerejeira et al., 2012; Sajjad et al., 2020). Hirsch et al. (2016) also showed that lower levels of IL-4, IL-5 and IL-12p70 were related to POD. IL-8 was analysed in three studies; in two of them, higher preoperative levels in CSF correlated with POD (Hirsch et al., 2016; Sajjad et al., 2020), but in one study, no differences were found (Cerejeira et al., 2012). Monocyte chemoattractant protein 1 (MCP-1) was analysed in two studies; one of them found that higher serum levels before surgery were associated with delirium (Kaźmierski et al., 2021), but the other one did not find any difference (Hirsch et al., 2016). Hirsch et al. (2016) also studied macrophage inflammatory protein (MIP), macrophage inflammatory protein (MRP), receptor for advanced glycation end products (RAGE) and calprotectin and found that POD was related to higher preoperative levels of MIP-1α MIP-1ß and calprotectin but not RAGE or MRP8/14. Preoperative chitinase 3-like 1 glycoprotein (CHI3L1) was evaluated in one study, but no association was found with delirium (Katsumi et al., 2020).

Human interferon-γ (IFN-γ) was investigated in two studies; one study found that lower preoperative IFN-γ in plasma was related to POD (Hirsch et al., 2016), but the other one did not find any differences in CSF and blood (Cape et al., 2014). Hirsch et al., 2016 also found that lower preoperative human interferon-α2 (IFN-α2) in CSF was implicated in the development of POD. Four studies explored insulin-like growth factor-1 (IGF-1). Two studies found that lower preoperative levels of IGF-1 were associated with POD (Çinar et al., 2014; Shen et al., 2016), but the other two studies did not find any differences (Cape et al., 2014; Chu et al., 2016). Glial fibrillary acidic protein (GFAP) was also investigated in 4 studies, but no association was found in any of them (Cape et al., 2014; Saller et al., 2019; Fong et al., 2020; Szwed et al., 2020). Only IL-6, TNF-α and CRP could be examined via meta-analysis, as each biomarker was investigated in 6 or more studies.

The major findings of this meta-analysis are presented in Figure 2. Overall, pooled analysis showed a significant increase in some serum biomarkers (i.e., CRP, TNF-α, and IL-6) of patients who developed delirium (OR = 1.88, 95% CI 1.01 to 1.637; I2 = 76.75%). Egger’s test indicated no small-study effects bias for pooled analysis (p = 0.178). Four studies could be included in the meta-analysis of IL-6; the results showed a significant increase in this biomarker in serum (OR = 1.88, 95% CI 1.01 to 1.637) in patients who developed delirium, with high heterogeneity (τ2 = 0.31, I2 = 76.75%). Four studies were included in the meta-analysis of CRP, but Dillon et al. (2017) evaluated three different cohorts with three independent analyses, so we included all of them in this meta-analysis. Serum preoperative CRP was significantly high in patients with later POD (OR = 1.75; 95% CI 1.04 to 2.93), with high heterogeneity (τ2 = 0.29, I2 = 72.92%).

FIGURE 2
www.frontiersin.org

Figure 2. Forest plot.

Biomarkers of dementia

Five studies investigated the relationship between biomarkers of dementia and delirium. Two studies analysed total tau (t-tau), but no differences were found between this biomarker and delirium in either blood (Fong et al., 2020) or CSF (Pan et al., 2019). Two studies evaluated phosphorylated tau (p-tau); in one study, higher CSF p-tau correlated with POD (Hov et al., 2017), but in another study, no differences were found between CSF p-tau and delirium (Pan et al., 2019). However, Saller et al. (2019) found that higher preoperative tau in serum correlated with POD. Hirsch et al. (2016) showed that lower preoperative Aβ40 in plasma predicted POD. Aß42 was investigated in two studies; one found that lower preoperative Aß42 in plasma correlated with incident POD (Hirsch et al., 2016), and the other found that lower preoperative Aß42 in CSF predicted POD (Pan et al., 2019).

Genetics

Only one study evaluated the implication of genetics in the development of delirium (Chen et al., 2020) showing that higher preoperative expression of miR-210 in blood was a predictor of POD.

Metabolomics, lipidomics and proteomics

Five studies explored the role of -omics approaches in delirium. Han et al., showed that CSF lipidomics and metabolomics pointed out that phosphatidylethanolamine (PE), phosphatydilcholine (PC), sphingomyelin (SM) and ceramide non-hydroxyfatty acid-sphingosine (Cer-NS) were related with delirium (Han et al., 2020b). In this study, lower preoperative levels of PE (40:7e), PE (40:6), PE (38:7e), PC (40:6) and PC (33:1) but higher preoperative levels of CER-NS and SM were associated with incident POD. A CSF proteomic study found that lower preoperative levels of transmembrane domain-containing protein 2B (VSTM2B) and coagulation factor V (FA5) were positively correlated with delirium severity (Han et al., 2020a). Vasunilashorn et al., showed that lower preoperative serum levels of zinc alpha-2 glycoprotein (AZGP1) were associated with POD (Vasunilashorn et al., 2019). Moreover, a serum proteomic study found that higher preoperative chitinase 3-like 1 glycoprotein (CHI3LI/KYL-40) were associated with POD (Vasunilashorn et al., 2021). Pan et al., showed that higher preoperative levels of spermidine, glutamine and putrescine in CSF were related with incident delirium (Pan et al., 2019).

Others

Bakker et al., showed that higher levels of serum creatinine prior to surgery were an independent predictor of POD (Bakker et al., 2012). Kotfis et al., found that lower preoperative levels of platelet-to-white blood cell ratio (PWR) and lower platelet-to-lymphocyte ratio (PLR) were associated with POD in serum (Kotfis et al., 2019). Szwed et al., investigated the relationship between neuroserpin (NSP) or visinin-like protein-1 (VILIP-1) and delirium (Szwed et al., 2020) in serum. Higher end of surgery to baseline ratio of NSP predicted the occurrence of POD but no differences were found between VILIP-1 and POD. Wyrobeck et al., showed that lower levels of serum Brain-derived neurotrophic factor (BDNF) during surgery were associated with delirium (Wyrobek et al., 2017).

Discussion

To the best of our knowledge, this is the first review that focuses on predictive biomarkers of delirium in older patients, considering all clinical settings and all types of biological fluids with high-quality studies. The multi-etiological nature of delirium is likely to be reflected in the wide range of biomarkers identified by our study.

Ach

The neurotransmitter hypothesis suggests that disturbances in neurotransmitter pathways can lead to delirium (Maldonado, 2013). Although precursors of serotonin (tryptophan, phenylalanine and tyrosine), dopamine and noradrenaline may be implicated in the development of delirium, anticholinergic deficiency has been directly related to its pathophysiology (Wang and Shen, 2018). Ach is involved in sleep regulation, cognition and attention. It is well known that an impairment in cholinergic activity is associated with cognitive and attentional changes; current literature also suggests that an impairment in cholinergic activity is associated with delirium (Pasina et al., 2019; Vondeling et al., 2020; Oudewortel et al., 2021; Lisibach et al., 2022). In addition, anticholinergic effects are common among several drugs, and a previous study found that increased anticholinergic burden increases delirium risk (Egberts et al., 2021).

Cortisol

A disruption of the hypothalamic–pituitary-axis with an increase in cortisol levels has been related to a higher risk of dementia (Ouanes and Popp, 2019) and delirium, particularly in critically ill patients (Mattar et al., 2012; Michels et al., 2019), but evidence is not conclusive. Other hormones, such as estradiol or leptin, may have a role in delirium pathophysiology, but additional research is needed to confirm this hypothesis.

S100B and NfL

Markers of neuronal damage, such as S100B and NfL (related to disturbances in astrocytic integrity), have been associated with delirium (Mietani et al., 2019, 2021), but these markers of brain injury could be associated with established, and not necessarily incident, delirium (Khan et al., 2011). However, higher levels have been associated with higher severity and worse prognosis of delirium (Gao et al., 2021; Narayanan et al., 2021; Page et al., 2022).

Tau and Aß

It is well known that cognitive impairment is a clinical risk factor for delirium (Fong et al., 2015, 2017, 2019; Racine et al., 2017). Nevertheless, few studies have been carried out in this field, pointing out that typical markers of dementia, such as tau or Aβ, are associated with delirium development (Wang et al., 2021, 2022).

Genetics

Another pathway that yields promising results is genetic research in delirium (Van Munster et al., 2009). Although studies in this field are scarce, genetic studies could be interesting translational tools for elucidating the pathophysiology of this syndrome. In addition, genetics are not influenced by the causal combination of predisposing and precipitating factors that affects delirium, so they can offer a more reliable value compared to other biomarkers (Sepulveda et al., 2021).

Proteomics and metabolomics

Research in proteomics and metabolomics aims to provide a protein or metabolic profile to contribute to delirium diagnosis and prevention (Sfera et al., 2015). There are few and heterogeneous studies in this field due to the technological difficulty associated with mapping the whole molecular landscape of biofluids. However, omics technologies are complementary to the genetic toolbox, detecting, identifying and quantifying alternative molecular biomarkers necessary to understand the dynamics and interactions that occur during delirium development.

Although acetylcholine deficiency, hormonal influence, markers of dementia and biochemical changes observed through proteomics and metabolomics may play an important role in predicting delirium, neuroinflammation theory seems to be the most relevant pathway, at least in the population of this review (most of the studies were carried out in older surgical populations). These findings are consistent with previous reviews that found an association between delirium and neuroinflammation biomarkers (Hall et al., 2018; Liu et al., 2018; Adamis et al., 2021; Dunne et al., 2021; Noah et al., 2021). This hypothesis suggests that peripheral inflammation due to surgery, trauma or infection leads to the activation of the proinflammatory cascade and suppression of anti-inflammatory markers. These stimuli trigger tissue macrophage and blood monocyte activation and secretion of inflammatory mediators such as IL-1, IL-1ß, IL-6, TNF-α and prostaglandin E2 (PGE2). These proinflammatory molecules penetrate the BBB, producing cerebral injury through the activation of microglia that causes brain dysfunction and delirium (Wilson et al., 2020). Interestingly, we conducted a meta-analysis with IL-6, TNF-α, and CRP, which were the most investigated biomarkers, and we found that they were statistically significant predictors of delirium.

Il-6

IL-6 is a cytokine involved in the immune response and has a notorious role in adult neurogenesis, the process of creating new neurons and glial cells from neural stem cells (oligodendrogliogenesis and astrogliogenesis) in the central nervous system (CNS) (Kang et al., 2020). IL-6 expression is involved in the synthesis of beta-amyloid precursor protein being altered in the brains of Alzheimer’s disease (AD) patients and it is also upregulated whenever neuroinflammation is expected, such as infection or injury (Erta et al., 2012). Noah et al. (2021) found that higher preoperative IL-6 was associated with postoperative delirium. Another meta-analysis performed among surgical patients also found this association (Liu et al., 2018; Adamis et al., 2021). Dunne et al. (2021) showed that early manifestation of systemic inflammation with elevated levels of IL-6 leads to the onset of delirium. However, Hall et al. (2018) did not find a significant correlation between IL-6 measured in CSF and delirium.

TNF-α

TNF-α is a proinflammatory cytokine (Idriss and Naismith, 2000) that has been associated with more rapid cognitive decline in patients with AD (Holmes et al., 2009). Analysis of the same cohort showed that elevated systemic TNF-α was associated with an increase in psychobehavioural alterations such as apathy, anxiety, depression and agitation, suggesting that increased systemic TNF-α may also have a role in hippocampal neurodegeneration (Holmes et al., 2011). The association of TNF-α and delirium is unclear. While some studies showed a positive correlation between TNF-α and delirium and our study revealed a significant association, previous meta-analyses by Liu et al. (2018) and Noah et al. (2021) found that preoperative TNF-α was significantly higher in the POD group in univariate analysis but not in multivariate analysis.

CRP

CRP is a pentameric protein whose circulating concentrations rise in response to inflammation (Pathak and Agrawal, 2019). It is an acute-phase protein of hepatic origin that increases following IL-6 secretion (Sproston and Ashworth, 2018). In the same way that IL-6 is altered in patients with cognitive impairment, a recent review found that CRP is also involved in this mechanism (Qiao et al., 2022). Moreover, several meta-analyses have shown that higher preoperative serum CRP levels are significantly associated with later POD (Liu et al., 2018; Adamis et al., 2021; Dunne et al., 2021; Noah et al., 2021).

Strengths and limitations

One of the main limitations of the studies included in this systematic review and meta-analysis is their small sample size and the lack of consistent terminology about delirium which has negatively affected the research (different terms such as organic brain syndrome, encephalopathy or acute brain failure can be misleading). Another important drawback is the lack of diversity in patient populations. Most of the studies were carried out in surgical populations, so their results may not be extrapolated to other clinical scenarios (medical inpatients, critically ill patients, etc.). On the other hand, there was little information about geriatric syndromes (frailty, malnutrition, polypharmacy) that may be directly involved not only in the development of delirium but also in the delirium characteristics (severity, subtype, duration, etc.). In addition, it is important to note that the methodological heterogeneity observed across studies might have an impact on the overall outcomes; therefore, the information should be considered with caution when drawing conclusions.

Importantly, this study provides several strengths. Firstly, there was no restriction by year of publication, and only high-quality studies (7 to 9 stars in NOS) with formal delirium diagnosis (DSM criteria, ICD criteria or a tool based on these criteria) with a lower risk of bias were included. Secondly, only studies on risk markers for delirium were included, excluding studies on disease markers or end-products of delirium. Thirdly, we also compared the preoperative state of patients who had not yet developed delirium in the studies that had more than one sample collected at different time points, focusing on predictive biomarkers of delirium and minimizing the risk of bias.

Challenges and implications for future research

These findings could contribute to the implementation of the International Drive to Illuminate Delirium (IDID) initiative (Khachaturian et al., 2020), providing a new prevention strategy for delirium and, subsequently, helping to preserve cognitive function. Although biomarkers could be an instrument to predict delirium, there are some unanswered questions. Blood-based biomarkers are little invasive, fast and reproducible, giving a quantitative value, and could be easy to measure in routine analysis (accessible in hands of surgeons, primary care physicians or other clinics). However, the validation of these biomarkers in terms of accuracy, sensitivity and specificity remains unknown because many of these inflammatory mediators are elevated in patients with sepsis, trauma or surgery and not only in those who develop delirium. In addition, their affordability and cost-efficiency are also unexplored.

Based on the limitations of the studies included in this systematic review and meta-analysis, we identified five major action areas to advance this research, designing better quality studies: (1) detailed planning and informed consent; (2) proper choice of target population; (3) blinding; (4) protocolization of biomarker collection and analysis; and (5) standardized reporting, which are summarized in Figure 3. and could be useful to improve the “Core Outcome Set” (COMET) in the field of delirium (Williamson et al., 2017).

FIGURE 3
www.frontiersin.org

Figure 3. Recommendations for future research in delirium biomarkers.

Conclusion

The relevance of delirium in older patients, both for its long- and short-term adverse consequences, provides a compelling reason to investigate its pathophysiology to prevent its appearance and improve the approach.

Despite the recent surge in studies on delirium biomarkers, there is not consistent evidence in the obtained results given the great heterogeneity that reflects the complexity of its pathophysiological mechanisms. Our data identified a significant association between biomarkers of neuroinflammation, such as CRP, IL-6, or TNF-α, and the risk of delirium, but further research is needed to standardize the methodology, improve scientific evidence and translate the current knowledge into pragmatic tools for routine clinical practice in the field of delirium.

Author contributions

LL-V, BC-V, and NM-V: conception and design of the work. LL-V and BC-V: acquisition of data. AG-H: analysis. AG-H and LL-V: interpretation of data. LL-V, NM-V, and RR-O: drafting of manuscript. JF-I, ES, FZ-F, MS, ÁM-V, and MI: critical revision. All authors contributed to the article and approved the submitted version.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fnagi.2023.1174644/full#supplementary-material

References

Abelha, F. J., Luís, C., Veiga, D., Parente, D., Fernandes, V., Santos, P., et al. (2013). Utcome and quality of life in patients with postoperative delirium during an ICU stay following major surgery. Crit. Care 17:R257. doi: 10.1186/cc13084

PubMed Abstract | CrossRef Full Text | Google Scholar

Adam, E. H., Haas, V., Lindau, S., Zacharowski, K., and Scheller, B. (2020). Cholinesterase alterations in delirium after cardiosurgery: a German monocentric prospective study. BMJ Open 10:e031212. doi: 10.1136/bmjopen-2019-031212

PubMed Abstract | CrossRef Full Text | Google Scholar

Adamis, D., van Gool, W. A., and Eikelenboom, P. (2021). Consistent patterns in the inconsistent associations of insulin-like growth factor 1 (IGF-1), C-reactive protein (C-RP) and interleukin 6 (IL-6) levels with delirium in surgical populations. A systematic review and meta-analysis. Arch. Gerontol. Geriatr. 97:104518. doi: 10.1016/j.archger.2021.104518

PubMed Abstract | CrossRef Full Text | Google Scholar

Altman, M. T., Knauert, M. P., Murphy, T. E., Ahasic, A. M., Chauhan, Z., and Pisani, M. A. (2018). Association of intensive care unit delirium with sleep disturbance and functional disability after critical illness: an observational cohort study. Ann. Intensive Care 8:63. doi: 10.1186/s13613-018-0408-4

CrossRef Full Text | Google Scholar

Aronson, J. K., and Ferner, R. E. (2017). Biomarkers—a general review. Curr. Protoc. Pharmacol. 76, 9.23.1–9.23.17. doi: 10.1002/cpph.19

CrossRef Full Text | Google Scholar

Avila-Funes, J. A., Ledesma-Heyer, J. P., Navarrete-Reyes, A. P., Chavira-Ramírez, R., Boeck-Quirasco, L., and Aguilar-Navarro, S. (2015). Association between high serum estradiol levels and delirium among hospitalized elderly women. Rev. Investig. Clin. organo del Hosp. Enfermedades la Nutr. 67, 20–24.

Google Scholar

Bakker, R. C., Osse, R. J., Tulen, J. H. M., Kappetein, A. P., and Bogers, A. J. J. C. (2012). Preoperative and operative predictors of delirium after cardiac surgery in elderly patients. Eur. J. cardio-thoracic Surg. Off. J. Eur. Assoc. Cardio-thoracic Surg. 41, 544–549. doi: 10.1093/ejcts/ezr031

CrossRef Full Text | Google Scholar

Cape, E., Hall, R. J., van Munster, B. C., de Vries, A., Howie, S. E. M., Pearson, A., et al. (2014). Cerebrospinal fluid markers of neuroinflammation in delirium: a role for interleukin-1β in delirium after hip fracture. J. Psychosom. Res. 77, 219–225. doi: 10.1016/j.jpsychores.2014.06.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Cavallari, M., Dai, W., Guttmann, C. R. G., Meier, D. S., Ngo, L. H., Hshieh, T. T., et al. (2016). Neural substrates of vulnerability to postsurgical delirium as revealed by presurgical diffusion MRI. Brain 139, 1282–1294. doi: 10.1093/brain/aww010

PubMed Abstract | CrossRef Full Text | Google Scholar

Cerejeira, J., Nogueira, V., Luís, P., Vaz-Serra, A., and Mukaetova-Ladinska, E. B. (2012). The cholinergic system and inflammation: common pathways in delirium pathophysiology. J. Am. Geriatr. Soc. 60, 669–675. doi: 10.1111/j.1532-5415.2011.03883.x

CrossRef Full Text | Google Scholar

Chen, Y., Lu, S., Wu, Y., Shen, Y., Zhao, H., Ding, S., et al. (2019). Change in serum level of interleukin 6 and delirium after coronary artery bypass graft. Am. J. Crit. care an Off. Publ. Am. Assoc. Crit. Nurses 28, 462–470. doi: 10.4037/ajcc2019976

CrossRef Full Text | Google Scholar

Chen, Y., Zheng, J., and Chen, J. (2020). Preoperative circulating MiR-210, a risk factor for postoperative delirium among elderly patients with gastric Cancer undergoing curative resection. Curr. Pharm. Des. 26, 5213–5219. doi: 10.2174/1381612826666200617163857

PubMed Abstract | CrossRef Full Text | Google Scholar

Choi, S. H., Lee, H., Chung, T. S., Park, K. M., Jung, Y. C., Kim, S. I., et al. (2012). Neural network functional connectivity during and after an episode of delirium. Am. J. Psychiatry 169, 498–507. doi: 10.1176/appi.ajp.2012.11060976

PubMed Abstract | CrossRef Full Text | Google Scholar

Chu, C.-L., Liang, C.-K., Lin, Y.-T., Chow, P. C., Pan, C.-C., Chou, M.-Y., et al. (2011). Biomarkers of delirium: well evidenced or not? J. Clin. Gerontol. Geriatr. 2, 100–104. doi: 10.1016/j.jcgg.2011.11.005

CrossRef Full Text | Google Scholar

Chu, C.-S., Liang, C.-K., Chou, M.-Y., Lin, Y.-T., Hsu, C.-J., Chu, C.-L., et al. (2016). Lack of association between pre-operative insulin-like growth Factor-1 and the risk of post-operative delirium in elderly Chinese patients. Psychiatry Investig. 13, 327–332. doi: 10.4306/pi.2016.13.3.327

PubMed Abstract | CrossRef Full Text | Google Scholar

Çinar, M. A., Balikçi, A., Sertoğlu, E., Mehmet, A. K., Serdar, M. A., and Özmenler, K. N. (2014). Role of CRP, TNF-a, and IGF-1 in delirium pathophysiology. Noro Psikiyatr. Ars. 51, 376–382. doi: 10.5152/npa.2014.6999

PubMed Abstract | CrossRef Full Text | Google Scholar

Cooper, H., Hedges, L. V., Valentine, J. C., and Rosenthal, R. E. (1994). Parametric Measures of Effect Size. The Handbook of Research synthesis. New York, NY: Russell Sage Foundation.

Google Scholar

de Jonghe, A., van Munster, B. C., Fekkes, D., van Oosten, H. E., and de Rooij, S. E. (2012). The tryptophan depletion theory in delirium: not confirmed in elderly hip fracture patients. Psychosomatics 53, 236–243. doi: 10.1016/j.psym.2011.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Dharmarajan, K., Swami, S., Gou, R. Y., Jones, R. N., and Inouye, S. K. (2017). Pathway from delirium to death: potential in-hospital mediators of excess mortality. J. Am. Geriatr. Soc. 65, 1026–1033. doi: 10.1111/jgs.14743

PubMed Abstract | CrossRef Full Text | Google Scholar

Dillon, S. T., Vasunilashorn, S. M., Ngo, L., Otu, H. H., Inouye, S. K., Jones, R. N., et al. (2017). Higher C-reactive protein levels predict postoperative delirium in older patients undergoing major elective surgery: a longitudinal nested case-control study. Biol. Psychiatry 81, 145–153. doi: 10.1016/j.biopsych.2016.03.2098

PubMed Abstract | CrossRef Full Text | Google Scholar

Dunne, S. S., Coffey, J. C., Konje, S., Gasior, S., Clancy, C. C., Gulati, G., et al. (2021). Biomarkers in delirium: a systematic review. J. Psychosom. Res. 147:110530. doi: 10.1016/j.jpsychores.2021.110530

PubMed Abstract | CrossRef Full Text | Google Scholar

Egberts, A., Moreno-Gonzalez, R., Alan, H., Ziere, G., and Mattace-Raso, F. U. S. (2021). Anticholinergic drug burden and delirium: a systematic review. J. Am. Med. Dir. Assoc. 22, 65–73.e4. doi: 10.1016/j.jamda.2020.04.019

CrossRef Full Text | Google Scholar

Egger, M., Smith, G. D., Schneider, M., and Minder, C. (1997). Bias in meta-analysis detected by a simple, graphical test. Br. Med. J. 315, 629–634. doi: 10.1136/bmj.315.7109.629

PubMed Abstract | CrossRef Full Text | Google Scholar

Erta, M., Quintana, A., and Hidalgo, J. (2012). Interleukin-6, a major cytokine in the central nervous system. Int. J. Biol. Sci. 8, 1254–1266. doi: 10.7150/ijbs.4679

PubMed Abstract | CrossRef Full Text | Google Scholar

Fong, T. G., Davis, D., Growdon, M. E., Albuquerque, A., and Inouye, S. K. (2015). The interface between delirium and dementia in elderly adults. Lancet Neurol. 14, 823–832. doi: 10.1016/S1474-4422(15)00101-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Fong, T. G., Inouye, S. K., and Jones, R. N. (2017). Delirium, dementia, and decline. JAMA Psychiat. 74, 212–213. doi: 10.1001/jamapsychiatry.2016.3812

CrossRef Full Text | Google Scholar

Fong, T. G., Vasunilashorn, S. M., Libermann, T., Marcantonio, E. R., and Inouye, S. K. (2019). Delirium and Alzheimer disease: a proposed model for shared pathophysiology. Int. J. Geriatr. Psychiatry 34, 781–789. doi: 10.1002/gps.5088

PubMed Abstract | CrossRef Full Text | Google Scholar

Fong, T. G., Vasunilashorn, S. M., Ngo, L., Libermann, T. A., Dillon, S. T., Schmitt, E. M., et al. (2020). Association of Plasma Neurofilament Light with postoperative delirium. Ann. Neurol. 88, 984–994. doi: 10.1002/ana.25889

PubMed Abstract | CrossRef Full Text | Google Scholar

Gao, Y., Duan, J., Ji, H., and Lu, W. (2021). Levels of S100 calcium binding protein B (S100B), neuron-specific enolase (NSE), and cyclophilin a (CypA) in the serum of patients with severe craniocerebral injury and multiple injuries combined with delirium transferred from the ICU and their prognosti. Ann. Palliat. Med. 10, 3371–3378. doi: 10.21037/apm-21-424

PubMed Abstract | CrossRef Full Text | Google Scholar

Gibb, K., Seeley, A., Quinn, T., Siddiqi, N., Shenkin, S., Rockwood, K., et al. (2020). The consistent burden in published estimates of delirium occurrence in medical inpatients over four decades: a systematic review and meta-analysis study. Age Ageing 49, 352–360. doi: 10.1093/ageing/afaa040

PubMed Abstract | CrossRef Full Text | Google Scholar

Gusmao-Flores, D., Figueira Salluh, J. I., Chalhub, R. T., and Quarantini, L. C. (2012). The confusion assessment method for the intensive care unit (CAM-ICU) and intensive care delirium screening checklist (ICDSC) for the diagnosis of delirium: a systematic review and meta-analysis of clinical studies. Crit. Care 16, R115–R111. doi: 10.1186/cc11407

PubMed Abstract | CrossRef Full Text | Google Scholar

Halaas, N. B., Zetterberg, H., Idland, A.-V., Knapskog, A.-B., Watne, L. O., and Blennow, K. (2021). Cerebrospinal fluid concentration of Neurogranin in hip fracture patients with delirium. J. Alzheimers Dis. 81, 667–677. doi: 10.3233/JAD-201341

PubMed Abstract | CrossRef Full Text | Google Scholar

Hall, R. J., Shenkin, S. D., and Maclullich, A. M. J. (2011). A systematic literature review of cerebrospinal fluid biomarkers in delirium. Dement. Geriatr. Cogn. Disord. 32, 79–93. doi: 10.1159/000330757

PubMed Abstract | CrossRef Full Text | Google Scholar

Hall, R. J., Watne, L. O., Cunningham, E., Zetterberg, H., Shenkin, S. D., Wyller, T. B., et al. (2018). CSF biomarkers in delirium: a systematic review. Int. J. Geriatr. Psychiatry 33, 1479–1500. doi: 10.1002/gps.4720

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, Y., Chen, W., Song, Y., Yuan, Y., Li, Z., Zhou, Y., et al. (2020a). Proteomic analysis of preoperative CSF reveals risk biomarkers of postoperative delirium. Front. Psych. 11:170. doi: 10.3389/fpsyt.2020.00170

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, Y., Zhang, W., Liu, J., Song, Y., Liu, T., Li, Z., et al. (2020b). Metabolomic and Lipidomic profiling of preoperative CSF in elderly hip fracture patients with postoperative delirium. Front. Aging Neurosci. 12:570210. doi: 10.3389/fnagi.2020.570210

PubMed Abstract | CrossRef Full Text | Google Scholar

Hayhurst, C. J., Marra, A., Han, J. H., Patel, M. B., Brummel, N. E., Thompson, J. L., et al. (2020). Association of Hypoactive and Hyperactive Delirium with cognitive function after critical illness. Crit. Care Med. 48, E480–E488. doi: 10.1097/CCM.0000000000004313

PubMed Abstract | CrossRef Full Text | Google Scholar

Helfand, B. K. I., D’Aquila, M. L., Tabloski, P., Erickson, K., Yue, J., Fong, T. G., et al. (2021). Detecting delirium: a systematic review of identification instruments for non-ICU settings. J. Am. Geriatr. Soc. 69, 547–555. doi: 10.1111/jgs.16879

PubMed Abstract | CrossRef Full Text | Google Scholar

Higgins, J. P., Thompson, S. G., Deeks, J. J., and Altman, D. G. (2003). Measuring inconsistency in meta-analyses. Br. Med. J. 327, 557–560. doi: 10.1136/bmj.315.7109.629

PubMed Abstract | CrossRef Full Text | Google Scholar

Hirsch, J., Vacas, S., Terrando, N., Yuan, M., Sands, L. P., Kramer, J., et al. (2016). Perioperative cerebrospinal fluid and plasma inflammatory markers after orthopedic surgery. J. Neuroinflammation 13:211. doi: 10.1186/s12974-016-0681-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Holmes, C., Cunningham, C., and Perry, V. H. (2009). Systemic inflammation and disease progression in Alzheimer disease. Neurology 73, 768–774. doi: 10.1212/WNL.0b013e3181d5df7f

PubMed Abstract | CrossRef Full Text | Google Scholar

Holmes, C., Cunningham, C., Zotova, E., Culliford, D., and Perry, V. H. (2011). Proinflammatory cytokines, sickness behavior, and Alzheimer disease. Neurology 77, 212–218. doi: 10.1212/WNL.0b013e318225ae07

PubMed Abstract | CrossRef Full Text | Google Scholar

Hov, K. R., Bolstad, N., Idland, A.-V., Zetterberg, H., Blennow, K., Chaudhry, F. A., et al. (2017). Cerebrospinal fluid S100B and Alzheimer’s disease biomarkers in hip fracture patients with delirium. Dement. Geriatr. Cogn. Dis. Extra 7, 374–385. doi: 10.1159/000481853

PubMed Abstract | CrossRef Full Text | Google Scholar

Ida, M., Takeshita, Y., and Kawaguchi, M. (2020). Preoperative serum biomarkers in the prediction of postoperative delirium following abdominal surgery. Geriatr Gerontol Int 20, 1208–1212. doi: 10.1111/ggi.14066

PubMed Abstract | CrossRef Full Text | Google Scholar

Idriss, H. T., and Naismith, J. H. (2000). TNFα and the TNF receptor superfamily: structure-function relationship(s). Microsc. Res. Tech. 50, 184–195. doi: 10.1002/1097-0029(20000801)50:3<184::AID-JEMT2>3.0.CO;2-H

PubMed Abstract | CrossRef Full Text | Google Scholar

Inouye, S. K., Van Dyck, C. H., Alessi, C. A., Balkin, S., Siegal, A. P., and Horwitz, R. I. (1990). Clarifying confusion: the confusion assessment method: a new method for detection of delirium. Ann. Intern. Med. 113, 941–948. doi: 10.7326/0003-4819-113-12-941

CrossRef Full Text | Google Scholar

Inouye, S. K., Westendorp, R. G. J., Saczynski, J. S., Naeije, G., and Pepersack, T. (2014). Delirium in elderly people. Lancet 383, 911–922. doi: 10.1016/S0140-6736(13)60688-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Israni, J., Lesser, A., Kent, T., and Ko, K. (2018). Delirium as a predictor of mortality in US Medicare beneficiaries discharged from the emergency department: a national claims-level analysis up to 12 months. BMJ Open 8, 1–7. doi: 10.1136/bmjopen-2017-021258

PubMed Abstract | CrossRef Full Text | Google Scholar

Kang, S., Narazaki, M., Metwally, H., and Kishimoto, T. (2020). Historical overview of the interleukin-6 family cytokine. J. Exp. Med. 217, 1–10. doi: 10.1084/jem.20190347

PubMed Abstract | CrossRef Full Text | Google Scholar

Katsumi, Y., Racine, A. M., Torrado-Carvajal, A., Loggia, M. L., Hooker, J. M., Greve, D. N., et al. (2020). The role of inflammation after surgery for elders (RISE) study: examination of [11C]PBR28 binding and exploration of its link to post-operative delirium. NeuroImage Clin. 27:102346. doi: 10.1016/j.nicl.2020.102346

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaźmierski, J., Miler, P., Pawlak, A., Jerczyńska, H., Woźniak, J., Frankowska, E., et al. (2021). Elevated monocyte Chemoattractant Protein-1 as the independent risk factor of delirium after cardiac surgery. A prospective cohort study. J. Clin. Med. 10:1587. doi: 10.3390/jcm10081587

PubMed Abstract | CrossRef Full Text | Google Scholar

Khachaturian, A. S., Hayden, K. M., Devlin, J. W., Fleisher, L. A., Lock, S. L., Cunningham, C., et al. (2020). International drive to illuminate delirium: a developing public health blueprint for action. Alzheimers Dement. 16, 711–725. doi: 10.1002/alz.12075

PubMed Abstract | CrossRef Full Text | Google Scholar

Khan, B. A., Zawahiri, M., Campbell, N. L., and Boustani, M. A. (2011). Biomarkers for deliriuma - A review. J. Am. Geriatr. Soc. 59, S256–S261. doi: 10.1111/j.1532-5415.2011.03702.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Kotfis, K., Ślozowska, J., Safranow, K., Szylińska, A., and Listewnik, M. (2019). The practical use of white cell inflammatory biomarkers in prediction of postoperative delirium after cardiac surgery. Brain Sci. 9:308. doi: 10.3390/brainsci9110308

PubMed Abstract | CrossRef Full Text | Google Scholar

Krewulak, K. D., Stelfox, H. T., Leigh, J. P., Wesley Ely, E., and Fiest, K. M. (2018). Incidence and prevalence of delirium subtypes in an adult ICU: a systematic review and meta-analysis. Crit. Care Med. 46, 2029–2035. doi: 10.1097/CCM.0000000000003402

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, G., Lei, X., Ai, C., Li, T., and Chen, Z. (2017). Low plasma leptin level at admission predicts delirium in critically ill patients: a prospective cohort study. Peptides 93, 27–32. doi: 10.1016/j.peptides.2017.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Liberati, A., Altman, D. G., Tetzlaff, J., Mulrow, C., Gøtzsche, P. C., Ioannidis, J. P. A., et al. (2009). The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. PLoS Med. 6:e1000100. doi: 10.1371/journal.pmed.1000100

PubMed Abstract | CrossRef Full Text | Google Scholar

Lisibach, A., Gallucci, G., Benelli, V., Kälin, R., Schulthess, S., Beeler, P. E., et al. (2022). Evaluation of the association of anticholinergic burden and delirium in older hospitalised patients – a cohort study comparing 19 anticholinergic burden scales. Br. J. Clin. Pharmacol. 88, 4915–4927. doi: 10.1111/bcp.15432

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, X., Yu, Y., and Zhu, S. (2018). Inflammatory markers in postoperativedelirium (POD) and cognitive dysfunction (POCD): a meta-analysis of observational studies. PLoS One 13:e0209284. doi: 10.1371/journal.pone.0195659

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, J. R., Fan, M. M., and Wang, Z. S. (2020). Age, preoperative higher serum cortisol levels, and lower serum acetylcholine levels predict delirium after percutaneous coronary intervention in acute coronary syndrome patients accompanied with renal dysfunction. Indian J. Psychiatry 62, 172–177. doi: 10.4103/psychiatry.IndianJPsychiatry_37_19

PubMed Abstract | CrossRef Full Text | Google Scholar

Maldonado, J. R. (2013). Neuropathogenesis of delirium: review of current etiologic theories and common pathways. Am. J. Geriatr. Psychiatry 21, 1190–1222. doi: 10.1016/j.jagp.2013.09.005

CrossRef Full Text | Google Scholar

Martínez-Velilla, N., Herrero, A. C., Cadore, E. L., Sáez de Asteasu, M. L., and Izquierdo, M. (2016). Iatrogenic nosocomial disability diagnosis and prevention. J. Am. Med. Dir. Assoc. 17, 762–764. doi: 10.1016/j.jamda.2016.05.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Mattar, I., Chan, M. F., and Childs, C. (2012). Factors causing acute delirium in critically ill adult patients: a systematic review. JBI Libr. Syst. Rev. 10, 187–231. doi: 10.11124/jbisrir-2012-3

CrossRef Full Text | Google Scholar

Michels, M., Michelon, C., Damásio, D., Vitali, A. M., Ritter, C., and Dal-Pizzol, F. (2019). Biomarker predictors of delirium in acutely ill patients: a systematic review. J. Geriatr. Psychiatry Neurol. 32, 119–136. doi: 10.1177/0891988719834346

PubMed Abstract | CrossRef Full Text | Google Scholar

Mietani, K., Hasegawa-Moriyama, M., Inoue, R., Ogata, T., Shimojo, N., Kurano, M., et al. (2021). Elevated neuron-specific enolase level is associated with postoperative delirium and detection of phosphorylated neurofilament heavy subunit: a prospective observational study. PLoS One 16, e0259217–e0259210. doi: 10.1371/journal.pone.0259217

PubMed Abstract | CrossRef Full Text | Google Scholar

Mietani, K., Sumitani, M., Ogata, T., Shimojo, N., Inoue, R., Abe, H., et al. (2019). Dysfunction of the blood-brain barrier in postoperative delirium patients, referring to the axonal damage biomarker phosphorylated neurofilament heavy subunit. PLoS One 14:e0222721. doi: 10.1371/journal.pone.0222721

PubMed Abstract | CrossRef Full Text | Google Scholar

Morandi, A., Rogers, B. P., Gunther, M. L., Merkle, K., Pandharipande, P., and Girard, T. D. (2012). The relationship between delirium duration, white matter integrity, and cognitive impairment in intensive care unit survivors as determined by diffusion tensor imaging: the VISIONS prospective cohort magnetic resonance imaging study. Crit. Care Med. 40, 2182–2189. doi: 10.1097/CCM.0b013e318250acdc.The

PubMed Abstract | CrossRef Full Text | Google Scholar

Murray, C., Sanderson, D. J., Barkus, C., Deacon, R. M. J., Rawlins, J. N. P., Bannerman, D. M., et al. (2012). Systemic inflammation induces acute working memory deficits in the primed brain: relevance for delirium. Neurobiol. Aging 33, 603–616.e3. doi: 10.1016/j.neurobiolaging.2010.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Narayanan, S., Shanker, A., Khera, T., and Subramaniam, B. (2021). Neurofilament light: a narrative review on biomarker utility. Fac. Rev. 10:46. doi: 10.12703/r/10-46

CrossRef Full Text | Google Scholar

Noah, A. M., Almghairbi, D., Evley, R., and Moppett, I. K. (2021). Preoperative inflammatory mediators and postoperative delirium: systematic review and meta-analysis. Br. J. Anaesth. 127, 424–434. doi: 10.1016/j.bja.2021.04.033

PubMed Abstract | CrossRef Full Text | Google Scholar

Osse, R. J., Fekkes, D., Tulen, J. H. M., Wierdsma, A. I., Bogers, A. J. J. C., van der Mast, R. C., et al. (2012). High preoperative plasma neopterin predicts delirium after cardiac surgery in older adults. J. Am. Geriatr. Soc. 60, 661–668. doi: 10.1111/j.1532-5415.2011.03885.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ouanes, S., and Popp, J. (2019). High cortisol and the risk of dementia and alzheimer’s disease: a review of the literature. Front. Aging Neurosci. 11:43. doi: 10.3389/fnagi.2019.00043

PubMed Abstract | CrossRef Full Text | Google Scholar

Oudewortel, L., van der Roest, H. G., Onder, G., Wijnen, V. J. M., Liperoti, R., Denkinger, M., et al. (2021). The Association of Anticholinergic Drugs and Delirium in nursing home patients with dementia: results from the SHELTER study. J. Am. Med. Dir. Assoc. 22, 2087–2092. doi: 10.1016/j.jamda.2021.05.039

PubMed Abstract | CrossRef Full Text | Google Scholar

Page, V. J., Watne, L. O., Heslegrave, A., Clark, A., McAuley, D. F., Sanders, R. D., et al. (2022). Plasma neurofilament light chain protein as a predictor of days in delirium and deep sedation, mortality and length of stay in critically ill patients. EBioMedicine 80, 104043–104012. doi: 10.1016/j.ebiom.2022.104043

PubMed Abstract | CrossRef Full Text | Google Scholar

Pan, X., Cunningham, E. L., Passmore, A. P., McGuinness, B., McAuley, D. F., Beverland, D., et al. (2019). Cerebrospinal fluid Spermidine, glutamine and Putrescine predict postoperative delirium following elective Orthopaedic surgery. Sci. Rep. 9:4191. doi: 10.1038/s41598-019-40544-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Pasina, L., Colzani, L., Cortesi, L., Tettamanti, M., Zambon, A., Nobili, A., et al. (2019). Relation between delirium and anticholinergic drug burden in a cohort of hospitalized older patients: an observational study. Drugs Aging 36, 85–91. doi: 10.1007/s40266-018-0612-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Pathak, A., and Agrawal, A. (2019). Evolution of C-reactive protein. Front. Immunol. 10:943. doi: 10.3389/fimmu.2019.00943

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, J., Wu, G., Chen, J., and Chen, H. (2019). Preoperative C-reactive protein/albumin ratio, a risk factor for postoperative delirium in elderly patients after Total joint Arthroplasty. J. Arthroplast. 34, 2601–2605. doi: 10.1016/j.arth.2019.06.042

PubMed Abstract | CrossRef Full Text | Google Scholar

Qiao, S., Li, H., Guo, F., Cai, G., and Zhang, Y. (2022). Research progress on cognitive impairment and the expression of serum inflammatory markers in patients with white matter hyperintensities: a narrative review. Ann. Transl. Med. 10:421. doi: 10.21037/atm-22-1016

PubMed Abstract | CrossRef Full Text | Google Scholar

Racine, A. M., Fong, T. G., Travison, T. G., Jones, R. N., Gou, Y., Vasunilashorn, S. M., et al. (2017). Alzheimer’s-related cortical atrophy is associated with postoperative delirium severity in persons without dementia. Neurobiol. Aging 59, 55–63. doi: 10.1016/j.neurobiolaging.2017.07.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Raichle, M. E. (2015). The Brain’s default mode network. Annu. Rev. Neurosci. 38, 433–447. doi: 10.1146/annurev-neuro-071013-014030

CrossRef Full Text | Google Scholar

Sachdev, P. S., Blacker, D., Blazer, D. G., Ganguli, M., Jeste, D. V., Paulsen, J. S., et al. (2014). Classifying neurocognitive disorders: the DSM-5 approach. Nat. Rev. Neurol. 10, 634–642. doi: 10.1038/nrneurol.2014.181

CrossRef Full Text | Google Scholar

Sajjad, M. U., Blennow, K., Knapskog, A. B., Idland, A.-V., Chaudhry, F. A., Wyller, T. B., et al. (2020). Cerebrospinal fluid levels of Interleukin-8 in delirium, dementia, and cognitively healthy patients. J. Alzheimers Dis. 73, 1363–1372. doi: 10.3233/JAD-190941

PubMed Abstract | CrossRef Full Text | Google Scholar

Saller, T., Petzold, A., Zetterberg, H., Kuhle, J., Chappell, D., von Dossow, V., et al. (2019). A case series on the value of tau and neurofilament protein levels to predict and detect delirium in cardiac surgery patients. Biomed. Pap. Med. Fac. Univ. Palacky, Olomouc, Czechoslov. 163, 241–246. doi: 10.5507/bp.2019.043

PubMed Abstract | CrossRef Full Text | Google Scholar

Sepulveda, E., Adamis, D., Franco, J. G., Meagher, D., Aranda, S., and Vilella, E. (2021). The complex interaction of genetics and delirium: a systematic review and meta-analysis. Eur. Arch. Psychiatry Clin. Neurosci. 271, 929–939. doi: 10.1007/s00406-021-01255-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Sfera, A., Price, A. I., Gradini, R., Cummings, M., and Osorio, C. (2015). Proteomic and epigenomic markers of sepsis-induced delirium (SID). Front. Mol. Biosci. 2:59. doi: 10.3389/fmolb.2015.00059

PubMed Abstract | CrossRef Full Text | Google Scholar

Shen, H., Shao, Y., Chen, J., and Guo, J. (2016). Insulin-like growth Factor-1, a potential predicative biomarker for postoperative delirium among elderly patients with open abdominal surgery. Curr. Pharm. Des. 22, 5879–5883. doi: 10.2174/1381612822666160813234311

PubMed Abstract | CrossRef Full Text | Google Scholar

Sillner, A. Y., Holle, C. L., and Rudolph, J. L. (2019). The overlap between falls and delirium in hospitalized older adults: a systematic review. Clin. Geriatr. Med. 35, 221–236. doi: 10.1016/j.cger.2019.01.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Sproston, N. R., and Ashworth, J. J. (2018). Role of C-reactive protein at sites of inflammation and infection. Front. Immunol. 9, 1–11. doi: 10.3389/fimmu.2018.00754

PubMed Abstract | CrossRef Full Text | Google Scholar

Strimbu, K., and Tavel, J. A. (2010). What are biomarkers? Curr. Opin. HIV AIDS 5, 463–466. doi: 10.1097/COH.0b013e32833ed177

PubMed Abstract | CrossRef Full Text | Google Scholar

Szwed, K., Słomka, A., Pawliszak, W., Szwed, M., Anisimowicz, L., Żekanowska, E., et al. (2020). Novel markers for predicting type 2 neurologic complications of coronary artery bypass grafting. Ann. Thorac. Surg. 110, 599–607. doi: 10.1016/j.athoracsur.2019.10.071

PubMed Abstract | CrossRef Full Text | Google Scholar

Trzepacz, P. T. (1999). The delirium rating scale. Its use in consultation-liaison research. Psychosomatics 40, 193–204. doi: 10.1016/S0033-3182(99)71235-1

CrossRef Full Text | Google Scholar

van Montfort, S. J. T., van Dellen, E., Stam, C. J., Ahmad, A. H., Mentink, L. J., Kraan, C. W., et al. (2019). Brain network disintegration as a final common pathway for delirium: a systematic review and qualitative meta-analysis. NeuroImage Clin. 23:101809. doi: 10.1016/j.nicl.2019.101809

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Munster, B. C., De Rooij, S. E., and Korevaar, J. C. (2009). The role of genetics in delirium in the elderly patient. Dement. Geriatr. Cogn. Disord. 28, 187–195. doi: 10.1159/000235796

PubMed Abstract | CrossRef Full Text | Google Scholar

Vasunilashorn, S. M., Dillon, S. T., Chan, N. Y., Fong, T. G., Joseph, M., Tripp, B., et al. (2021). Proteome-wide analysis using SOMAscan identifies and validates Chitinase-3-like protein 1 as a risk and disease marker of delirium among older adults undergoing major elective surgery. J. Gerontol. A Biol. Sci. Med. Sci. 77, 484–493. doi: 10.1093/gerona/glaa326

PubMed Abstract | CrossRef Full Text | Google Scholar

Vasunilashorn, S. M., Ngo, L. H., Chan, N. Y., Zhou, W., Dillon, S. T., Otu, H. H., et al. (2019). Development of a dynamic multi-protein signature of postoperative delirium. J. Gerontol. A Biol. Sci. Med. Sci. 74, 261–268. doi: 10.1093/gerona/gly036

PubMed Abstract | CrossRef Full Text | Google Scholar

Vondeling, A. M., Knol, W., Egberts, T. C. G., and Slooter, A. J. C. (2020). Anticholinergic drug exposure at intensive care unit admission affects the occurrence of delirium. A prospective cohort study. Eur. J. Intern. Med. 78, 121–126. doi: 10.1016/j.ejim.2020.04.062

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S., Greene, R., Song, Y., Chan, C., Lindroth, H., Khan, S., et al. (2022). Postoperative delirium and its relationship with biomarkers for dementia: a meta-analysis. Int. Psychogeriatrics 34, 377–390. doi: 10.1017/S104161022100274X

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S., Lindroth, H., Chan, C., Greene, R., Serrano-Andrews, P., Khan, S., et al. (2021). A systematic review of delirium biomarkers and their alignment with the NIA-AA research framework. J. Am. Geriatr. Soc. 69, 255–263. doi: 10.1111/jgs.16836

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y., and Shen, X. (2018). Postoperative delirium in the elderly: the potential neuropathogenesis. Aging Clin. Exp. Res. 30, 1287–1295. doi: 10.1007/s40520-018-1008-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Watt, C. L., Momoli, F., Ansari, M. T., Sikora, L., Bush, S. H., Hosie, A., et al. (2019). The incidence and prevalence of delirium across palliative care settings: a systematic review. Palliat. Med. 33, 865–877. doi: 10.1177/0269216319854944

PubMed Abstract | CrossRef Full Text | Google Scholar

Welch, C., McCluskey, L., Wilson, D., Chapman, G. E., Jackson, T. A., Treml, J., et al. (2019). Delirium is prevalent in older hospital inpatients and associated with adverse outcomes: results of a prospective multi-Centre study on world delirium awareness day. BMC Med. 17, 1–11. doi: 10.1186/s12916-019-1458-7

CrossRef Full Text | Google Scholar

Williamson, P. R., Altman, D. G., Bagley, H., Barnes, K. L., Blazeby, J. M., Brookes, S. T., et al. (2017). The COMET handbook: version 1.0. Trials 18, 1–50. doi: 10.1186/s13063-017-1978-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Wilson, J. E., Mart, M. F., Cunningham, C., Shehabi, Y., Girard, T. D., Maclullich, A. M. J., et al. (2020). Delirium. Nature 11. doi: 10.1038/s41572-020-00223-4

CrossRef Full Text | Google Scholar

World Health Organization (WHO) (2001). Biomarkers in Risk Assessment: Validity and Validation. World Health Organization, Geneva, 144.

Google Scholar

Wyrobek, J., LaFlam, A., Max, L., Tian, J., Neufeld, K. J., Kebaish, K. M., et al. (2017). Association of intraoperative changes in brain-derived neurotrophic factor and postoperative delirium in older adults. Br. J. Anaesth. 119, 324–332. doi: 10.1093/bja/aex103

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: delirium, cognitive impairment, biomarkers, neuroinflammation, older people

Citation: Lozano-Vicario L, García-Hermoso A, Cedeno-Veloz BA, Fernández-Irigoyen J, Santamaría E, Romero-Ortuno R, Zambom-Ferraresi F, Sáez de Asteasu ML, Muñoz-Vázquez ÁJ, Izquierdo M and Martínez-Velilla N (2023) Biomarkers of delirium risk in older adults: a systematic review and meta-analysis. Front. Aging Neurosci. 15:1174644. doi: 10.3389/fnagi.2023.1174644

Received: 26 February 2023; Accepted: 26 April 2023;
Published: 12 May 2023.

Edited by:

Guoyuan Qi, University of Arizona, United States

Reviewed by:

Christian Barbato, National Research Council (CNR), Italy
Leda Leme Talib, University of São Paulo, Brazil

Copyright © 2023 Lozano-Vicario, García-Hermoso, Cedeno-Veloz, Fernández-Irigoyen, Santamaría, Romero-Ortuno, Zambom-Ferraresi, Sáez de Asteasu, Muñoz-Vázquez, Izquierdo and Martínez-Velilla. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Lozano-Vicario Lucía, lucia.lozanovicario@gmail.com

Download