Skip to main content
Sage Choice logoLink to Sage Choice
. 2021 Jun 26;35(8):934–945. doi: 10.1177/02698811211026426

Ketamine’s effect on inflammation and kynurenine pathway in depression: A systematic review

Emma Kopra 1, Valeria Mondelli 1,2, Carmine Pariante 1,2, Naghmeh Nikkheslat 1,
PMCID: PMC8358579  PMID: 34180293

Abstract

Background:

Ketamine is a novel rapid-acting antidepressant with high efficacy in treatment-resistant patients. Its exact therapeutic mechanisms of action are unclear; however, in recent years its anti-inflammatory properties and subsequent downstream effects on tryptophan (TRP) metabolism have sparked research interest.

Aim:

This systematic review examined the effect of ketamine on inflammatory markers and TRP–kynurenine (KYN) pathway metabolites in patients with unipolar and bipolar depression and in animal models of depression.

Methods:

MEDLINE, Embase, and PsycINFO databases were searched on October 2020 (1806 to 2020).

Results:

Out of 807 initial results, nine human studies and 22 animal studies on rodents met the inclusion criteria. Rodent studies provided strong support for ketamine-induced decreases in pro-inflammatory cytokines, namely in interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α and indicated anti-inflammatory effects on TRP metabolism, including decreases in the enzyme indoleamine 2,3-dioxygenase (IDO). Clinical evidence was less robust with high heterogeneity between sample characteristics, but most experiments demonstrated decreases in peripheral inflammation including in IL-1β, IL-6, and TNF-α. Preliminary support was also found for reduced activation of the neurotoxic arm of the KYN pathway.

Conclusion:

Ketamine appears to induce anti-inflammatory effects in at least a proportion of depressed patients. Suggestions for future research include investigation of markers in the central nervous system and examination of clinical relevance of inflammatory changes.

Keywords: Ketamine, inflammation, cytokine, kynurenine, depression

Introduction

Major depressive disorder (MDD) is among the leading causes of disability worldwide, contributing to high human and societal costs (Ferrari et al., 2013). For decades, depression has primarily been treated with antidepressants targeting monoamine function; however, about one-third of patients are considered treatment-resistant to such medications, lacking adequate response to two or more antidepressant trials (Nemeroff, 2007). For those who respond, effects take usually 2–4 weeks to be noticeable and a large proportion eventually relapse (Gaynes et al., 2009). In bipolar disorder (BD), where depressive episodes alternate with manic episodes and antidepressant response rates are even lower (Pacchiarotti et al., 2013) with fewer available treatment options than in MDD (Vieta et al., 2010). Novel treatments and drug targets are therefore urgently needed.

Ketamine, a noncompetitive antagonist of the glutamate N-methyl-D-aspartate (NMDA) receptor, has been referred to as one of the most important discoveries in antidepressant research in half a century (Duman and Aghajanian, 2012). It has been used as a dissociative anesthetic since the 1960s, but its therapeutic potential was not discovered until the year 2000 (Berman et al., 2000). Since then, numerous studies have replicated ketamine’s antidepressant and anti-suicidal effects in both unipolar and bipolar depression (Coyle and Laws, 2015; Wilkinson et al., 2018) with high response rates of up to 60%–70% in treatment-resistant patients (Bobo et al., 2016; Diazgranados et al., 2010; Murrough et al., 2013a). Ketamine is fast acting with effects emerging within hours (Bobo et al., 2016) making it a beneficial tool, especially for patients in acute risk of self-harm and suicide (Larkin and Beautrais, 2011). Ketamine’s effects peak approximately 1-day post-infusion and last around 1 week (Corriger and Pickering, 2019; Kishimoto et al., 2016) but can be prolonged with repeated infusions (Murrough et al., 2013b; Zheng et al., 2018). The (S)-isomer of ketamine (esketamine) was approved as a medication for treatment-resistant MDD in nasal spray form in the United States in 2019 (FDA, 2019).

The exact mechanisms behind ketamine’s antidepressant effects are yet to be fully understood. However, its NMDA receptor antagonist property has been postulated to play a central role, the therapeutic mechanisms are believed to be much more complex. This is supported by the existence of several other NMDA receptor antagonists without antidepressant properties (Newport et al., 2015), as well as preclinical research on the pharmacology and efficacy of ketamine’s enantiomers (Hashimoto, 2019; Jelen et al., 2020; Yang et al., 2015a) and metabolites (Hashimoto, 2019; Zanos et al., 2016). In recent years, there has been an increasing interest in ketamine’s anti-inflammatory properties. Ketamine has been shown to attenuate heightened inflammation in animal models, in human blood in vitro (De Kock et al., 2013; Loix et al., 2011), and in surgical contexts when used as an anesthetic or antihyperalgesic (Dale et al., 2012; Loix et al., 2011).

The anti-inflammatory effects of ketamine have been of particular interest given that the association between inflammation and depression is well-established. This relationship is likely to be bidirectional, with high inflammation levels (Khandaker et al., 2014; Valkanova et al., 2013) and presence of inflammation-related diseases (Benros et al., 2013; Sforzini et al., 2019; Wang et al., 2018) increasing not only the risk of developing MDD and BD, but also vice versa (Andersson et al., 2015; Rosenblat and McIntyre, 2017). Increased inflammatory markers, specifically C-reactive protein (CRP), interleukin (IL)-1, IL-6, and tumor-necrosis factor alpha (TNF-α), have been observed in the blood and cerebrospinal fluid (CSF) as well as in postmortem brain samples of MDD and BD patients (Enache et al., 2019; Osimo et al., 2020; Raison et al., 2006; Rosenblat et al., 2014). Interestingly, inflammation is prevalent, especially in treatment-resistant depression (Cattaneo et al., 2020; Strawbridge et al., 2015; Yang et al., 2019); and longitudinal investigations have found patients with higher baseline inflammation to be less likely to respond to traditional antidepressants (Cattaneo et al., 2013, 2016; Strawbridge et al., 2015).

Among the main mechanisms, inflammation is suggested to induce depressive symptoms through its effects on the tryptophan (TRP)–kynurenine (KYN) pathway, as supported by studies in which activation of KYN pathway and increased depressive symptoms were observed in patients with hepatitis C virus infection undergoing interferon (IFN)-α treatment (Raison et al., 2010). Indeed, pro-inflammatory cytokines IFN-γ, IFN-α, IL-1, and TNF-α activate the enzyme indoleamine 2,3-dioxygenase (IDO), which metabolizes TRP into KYN instead of serotonin (Maes et al., 2007). The pro-inflammatory cytokine IL-1β further activates the enzyme 3-monooxygenase (KMO; Zunszain et al., 2012), which converts KYN into its neurotoxic microglial by-products, such as 3-hydroxykynurenine (3-HK), 3-hydroxyanthranilic acid (3-HAA) and eventually quinolinic acid (QUIN; Ogyu et al., 2018; Parrott et al., 2016). These metabolites contribute to neurotoxicity through numerous mechanisms and can also further exacerbate inflammation. For instance, QUIN agonizes the NMDA receptor inducing glutamate excitotoxicity and contributing to a signaling cascade that leads to reduced brain-derived neurotropic factor (BDNF), protein synthesis, and synaptogenesis (Lugo-Huitrón et al., 2013), whereas 3-HK increases reactive oxygen species in the brain, contributing to oxidative stress and neuronal apoptosis, especially in the hippocampus (Colín-González et al., 2013). Under normal conditions, KYN is mainly metabolized in astrocytes by kynurenine aminotransferases (KATs) to kynurenic acid (KynA). KynA is an antagonist of the NMDA receptor and to a lesser extent the α7-nicotinic acetylcholine and AMPA receptors, through which it exerts neuroprotective and anti-inflammatory effects, enhances synaptic plasticity, and clears excess glutamate in the brain (Ganong and Cotman, 1986; Potter et al., 2010).

Evidence is rapidly growing on the role of inflammation in association with KYN pathway abnormalities and dysregulation of KYN metabolites in patients who have committed suicide (Bryleva and Brundin, 2017). However, the data is less robust for the whole MDD population. A recent systematic review observed decreased KYN and KynA in depressed patients, alongside increased QUIN in antidepressant-free patients, compared to healthy controls (Ogyu et al., 2018); however, some studies have found no differences between KYN pathway metabolites of depressed patients and healthy controls in peripheral (Hughes et al., 2012) nor central samples (Hestad et al., 2017; Miller et al., 2008). Decreased TRP among patients with no changes in KYN or its downstream components has been also observed (Gabbay et al., 2010; Hughes et al., 2012), suggesting potential involvement of alternative mechanisms in the depletion of TRP.

Understanding ketamine’s potential effect on inflammatory proteins and TRP metabolism would increase our understanding of the biological mechanisms behind its antidepressant effect and help identify pharmacological targets for other antidepressants. Crucially, understanding ketamine’s immunomodulatory mechanisms could inform us of potential predictors of response and therefore help reduce unnecessary treatment trials. Changes in cytokines have previously been found to be associated with antidepressant response to traditional antidepressants (Cattaneo et al., 2013; Lanquillon et al., 2000), and a recent systematic review concluded that treatment-resistant patients with higher inflammation have better response to medication with anti-inflammatory characteristics including ketamine (Yang et al., 2019). Identification of predictors of response as well as development of novel pharmacotherapies are of particular interest given concerns surrounding ketamine’s side-effects and potential risks of long-term use (Short et al., 2018).

Thus far research on the effect of ketamine on inflammation has yielded mixed reports with heterogeneity in methodologies and samples complicating evidence synthesis. The aim of this systematic review is to analyze current evidence on the effect of ketamine on inflammatory markers and TRP–KYN pathway metabolites in unipolar and bipolar depression, as well as in preclinical studies employing animal models of depression.

Methods

Search strategy

Electronic literature databases MEDLINE ([R] and Epub Ahead of Print, In-Process, In-Data-Review & Other Non-Indexed Citations and Daily; 1946 to October 4, 2020), APA PsycInfo (1806 to September Week 4, 2020), and Embase (1974 to 2020 Week 40) were searched through Ovid interface (https://ovidsp.ovid.com/) to find relevant studies. The following combination of keywords was entered: Ketamine AND Depress* AND Inflammat*/cytokine*/interleukin/c-reactive protein/CRP/tumor necrosis factor*/TNF*/interferon/IFN*/kynuren*/KYNA/quinolinic/QUIN/QA. Results were limited to publications in English language. No restrictions were placed regarding publication year. Reference sections of included studies and review articles on the topic were screened to identify additional relevant publications. The final search was performed on October 5th, 2020.

Selection of literature

References were imported into the RefWorks citation manager tool (ProQuest, Ann Arbor, MI, USA) for screening of results. Participants in human studies were required to have a current Diagnostic and Statistical Manual of Mental Disorders (DSM) or International Classification of Diseases (ICD) diagnosis of either MDD or BD and to be experiencing a depressive episode at the time of the study. Animal studies were required to use established animal models of depression such as lipopolysaccharide- (LPS) or stress-induced depression.

Studies were required to include in vivo administration of ketamine. Studies were excluded if ketamine was administered as an adjunct therapy with another pharmacological or psychological treatment, though participants were allowed to be maintained on their current medications. Human studies were required to measure biomarkers at baseline and at least once posttreatment and provide a within-subject analysis of the change, or at minimum provide sufficient data for interpretation of this change. Animal studies were required to include comparison of posttreatment biomarkers between ketamine group and a control group with otherwise identical treatment but without administration of an active drug.

Quality assessment

The quality of human studies was evaluated with the following parameters: (1) blinding of patients and investigators, (2) absence of adjunct medication, (3) number of outcome measurement points, (4) appropriate statistical analyses, and (5) completeness of data and reporting. Animal studies’ quality was assessed based on (1) depression model, (2) clarity of protocol and similar treatment of groups, (3) appropriate statistical analyses; and (4) completeness of data and reporting. Each parameter was awarded a maximum score of two, depending on how well each study fulfilled the criterion (0 = low/absent, 1 = partial, and 2 = full). A total quality score was generated by summing the scores for all parameters, with a maximum score of 10 for human studies and 8 for animal studies.

Results

Study selection

The initial search yielded 848 results, reducing to 807 after limiting results to English language and 581 after deduplication. About 62 articles were selected for full-text assessment after the initial title and abstract screening, of which 31 were included in the qualitative analysis. Nine of the included studies were on humans and 22 on rodents. A flow diagram of the search process with reasons for exclusions is presented in Supplemental Figure S1.

Description of selected studies

Human studies

A summary of human study characteristics is shown in Table 1. All studies were published between 2015 and 2020. Seven studies measured inflammatory markers, the most widely measured being IL-6 (n = 7) and TNF-α (n = 6); and five studies measured TRP–KYN metabolites, the most commonly measured being KYN (n = 5) and TRP (n = 4). All studies measured protein levels of the markers. The samples of studies by Kadriu et al. (2019) and Park et al. (2017) overlapped, and therefore for the former we will only report results for KYN metabolites but not for cytokines, which are analyzed more thoroughly and in a larger sample by Park et al. (2017).

Table 1.

Study characteristics and results of human studies.

Study Sample Medication Design Intervention Outcome measures Findings
Allen et al. (2018) 17 TRD 15/17 (88%) Medicated Open label, repeated measures 0.5 mg/kg i.v. ketamine once a week for 3 weeks (=3 infusions) Whole blood IL-6, IL-8, IL-10, IFN-γ; and KYN, KynA, TRP, KYN/TRP ratio, KynA/KYN ratio; at baseline, 2 h and 1-week post each infusion, and 24 h post-first infusion No significant changes after ketamine
 Ireland 47% Female patients In responders (HDRS reduction >50%; n = 13), trend towards decreased KYN 2 h after first infusion (p = 0.067), and reduced KYN/TRP ratio at 24 h after first infusion (p = 0.054)
Chen et al. (2018) 71 TRD (n = 23–24/group) Medications continued (no numbers given) Double-blind RCT Single 0.5 mg/kg i.v. ketamine; 0.2 mg/kg i.v. ketamine; or i.v. saline Serum CRP, IL-6, and TNF-α at baseline, 40 min, 240 min, day 3, and day 7 post-infusion IL-6 and TNF-α levels differed significantly over time in all groups (p = 0.002, p = 0.001). TNF-α was significantly lower at 40 and 240 min post-infusion than at baseline for the 0.5 mg/kg group (p < 0.05)
 Taiwan 75% Female patients
Kadriu et al. (2019) a 39 TRBD All patients on mood stabilizers only Sample drawn from a randomized placebo-controlled crossover study Single 0.5 mg/kg i.v. ketamine over 40 min Plasma IDO, KYN, KynA, and QA at baseline, 230 min, day 1 and day 3 post-infusion. IDO levels significantly lower than baseline at all three time points, t(34) = −3.05, p = 0.004; t(34) = −4.12, p = 0.0002; t(34) = −3.91, p = 0.0004 for 230 min, 1 day, and 3 days, respectively.
 USA 59% Female patients KYN significantly increased at 1 day (t = 3.69, p = 0.0008) and 3 day (t = 2.44, p = 0.02). KynA significantly increased at 1 day (t = 2.85, p = 0.007) and 3 day (t = 3.36, p = 0.002). KYN/KynA ratio significantly increased (t = 2.91, p = 0.006) and QA/KYN ratio significantly decreased (t = −3.31, p = 0.002) at day 1.
Kiraly et al. (2017) 33 TRD Free of medication for ⩾2 weeks Open label, repeated measures Single 0.5 mg/kg i.v. ketamine over 40 min Serum levels of 41 cytokines, chemokines and growth factors at baseline; and 4 h and 24 h post-infusion Significant decreases in IL-6 (t = 2.369, p < 0.05), IL-1α (p < 0.05, t = 2.149), IL-13 (p = 0.038), G-CSF (p = 0.038), and IP-10/CXCL10 (p < 0.0001) at 4 h; alongside decreases in IL-8 (p < 0.0001) and PDGF-AA (p = 0.024); and increases in IL-7 (p < 0.0001) at 24 h
 USA
Moaddel et al. (2018) 29 TRDb Free of medication for ⩾2 weeks Double-blind, placebo-controlled crossover study Single 0.5 mg/kg i.v. ketamine or saline over 40 mins, 2 weeks apart Plasma KYN and TRP at baseline, 40 min (end of infusion), 80 min, 110 min, 230 min, and at days 1, 2 and 3 post-infusion No significant changes after ketamine
 USA When compared to placebo, changes in KYN/TRP ratio (p = 0.013) and KYN (p = 0.039), namely a slight initial increase in ketamine group compared to a sharp drop in placebo group
Park et al. (2017) a 49 TRD and 31 TRBD TRD free of medication for ⩾2 weeks; BD on mood stabilizers only Sample drawn from double-blind placebo-controlled studies Single 0.5 mg/kg i.v. ketamine over 40 mins Plasma IL-2, IL-5, IL-6, IL-8, IL-10, IFN-γ, TNF-α and sTNFR1 at baseline and at 230 min, 1 day and 3 days post-infusion Significantly increased IL-6 (F3,209 = 25.51, p < 0.001) and decreased sTNFR1 (F3,206 = 4.27, p = 0.006) 230 min post-infusion
 USA 51% Female patients
Yang et al. (2015b) 16 TRDc Free of medication for ⩾2 weeks Open label, repeated measures Single 0.5 mg/kg i.v. ketamine over 40 mins Serum IL-1β, IL-6, and TNF-α; and KYN and TRP; at baseline, 230 mins and 1- and 3-day post-infusion In the responder group (MADRS reduction >50%; n = 12), IL-1β showed significant decrease at 230 mins and 1-day post-infusion (F = 4.495, df = 2.602, p = 0.013), and IL-6 significant decrease at 230 mins to 3 days post-infusion (F = 9.450, df = 2.914, p < 0.001)
 China/Japan
Zhan et al. (2020) 60 MDD (TR or suicidal) All medicated Open label, repeated measures Six 0.5 mg/kg i.v. ketamine infusions (40 mins) over a 12-day period Plasma levels of 19 cytokines at baseline, 24 h after first infusion, and 24 h and 14 days after sixth infusion Levels of 14 cytokines significantly decreased at 14 days post-sixth infusion; IL-1β, IL-4, IL-5, IL-12p70, IL-23, and GM-CSF (p < 0.05); and IL-2, IL-6, IL-7, IL-10, IL-17A, IFN-γ, TNF-α, and fractalkine (p < 0.01)
 China 63% Female patients
Zhou et al. (2018) 68 MDD and 16 BD (TR = 73; suicidal = 48) All medicated Open label, repeated measures Six 0.5 mg/kg i.v. ketamine infusions (40 min) over a 12-day period Serum TRP, KYN, and KynA at baseline, 24 h after first infusion, and 24 h and 14 days after sixth infusion In responders (MADRS reduction >50%; n = 50), KynA and KynA/KYN ratio increased at 24 h after first infusion (KynA: t = 2.842, p = 0.031; KynA/KYN ratio: t = 2.842, p = 0.031) and 24 h after sixth infusion (KynA: t = −2.946, p = 0.023; KynA/KYN ratio: t = −2.919, p = 0.025)
 China 54% Female patients

3-HK: 3-hydroxykynurenine; 3-HAA: 3-hydroxyanthranilic acid; BD: bipolar disorder; CRP: C-reactive protein; CSF: cerebrospinal fluid; G-CSF: granulocyte colony-stimulating factor; GM-CSF: granulocyte-macrophage colony-stimulating factor; HDRS: Hamilton depression rating scale; IDO: indoleamine 2,3-dioxygenase; IL: interleukin; IFN-α: interferon-alpha; i.v.: intravenous; KYN: kynurenine; KynA: kynurenic acid ; MDD: major depressive disorder; MADRS: Montgomery–Åsberg depression rating scale; TRBD: treatment-resistant bipolar depression; TNF-α: tumor necrosis factor-alpha; TRD: treatment-resistant major depression; TRP: tryptophan.

a

Kadriu et al. (2019) and Park et al. (2017) had partially overlapping sample; therefore, cytokine data only reported for Park et al (2017).

b

TR criteria required only one failed previous antidepressant trial.

c

TR criteria not specified.

The number of patients receiving ketamine ranged from 16 to 84, the combined sample of all included studies totaling 429. Six studies included MDD patients, one BD patients, and two studies included both. Most studies employed treatment-resistant criteria requiring patients to have had at least two previous failed antidepressant trials, except from one study only requiring a single failed trial (Moaddel et al., 2018). Two studies allowed inclusion of nontreatment-resistant patients if these were suicidal (Zhan et al., 2020; Zhou et al., 2018). In four out of six studies, MDD patients were medication free, and in two out of three studies, BD patients were allowed mood stabilizers only. Six studies administered ketamine once, and three had multiple infusions. All studies used a 0.5 mg/kg intravenous dose of ketamine with one study also including a 0.2 mg/kg condition (Chen et al., 2018). Studies included between 2 and 7 post-infusion measurements, from immediately after administration to up to 2-week post-infusion.

Study quality scoring is shown in Supplemental Table S1. Total scores ranged between four and eight out of ten. Statistical analyses were appropriate by most part; however, a few studies did exploratory analyses with several markers without adjusting for multiple comparisons where this would have been considered appropriate. Moaddel et al. (2018) did not conduct a simple within-subject analysis of ketamine-induced changes but rather contrasted effects to a placebo group; therefore, absolute changes in levels had to be interpreted from a graph. Two studies provided only stratified data according to antidepressant responder status.

Animal studies

Animal study characteristics are summarized in Table 2. All studies were published between 2013 and 2020. Sixteen studies examined inflammatory markers, one study TRP– KYN pathway metabolites, and five studies measured both. The most measured markers were IL-1β and IL-6, measured in 19 and 18 studies, respectively. All studies measured protein levels of the markers, except from three studies measuring mRNA levels of cytokines and one study measuring mRNA levels of IDO. Twelve studies measured markers in either the brain (most commonly hippocampus or prefrontal cortex) or CSF, seven in blood, and three studies reported levels for both. In 13 studies, tissue or blood samples were taken less than 24 h from treatment, in seven studies 24 h or more, and two did both with different groups of rodents. Ketamine was delivered intraperitoneally with doses ranging from 5 to 20 mg/kg, except from one study with a 100 mg/kg dose (Zhu et al., 2015) and one with an additional 90 mg/kg condition besides a lower-dose group (Verdonk et al., 2019). About 17 studies administered a single ketamine infusion, three injected multiple infusions, and two included both conditions.

Table 2.

Study characteristics and results of animal studies.

Study Animalsa Model Intervention Outcome measuresb Findings
Abelaira et al. (2017) 8–10 Adult male Wistar rats FST Ketamine 15 mg/kg i.p. or saline Serum IL-1β, IL-10, and TNF-α TNF-α increased in ketamine group (p < 0.05)
0.5 h
Aricioglu et al. (2020) 30 Adult male Wistar albino rats CUMS for 6 weeks Ketamine 10 mg/kg i.p. single dose, or daily for 3 weeks (in the last 3 weeks of CUMS), or saline PFC IL-1β and IL-6 (mRNA) Both acute and chronic ketamine decreased IL-1β; only acute ketamine decreased IL-6 (p < 0.05)
7 days
Chang et al. (2019) 20 Adult male Wistar rats LPS Ketamine 10 mg/kg, i.p. or saline Hippocampal tissue IL-1β, IL-6, and TNF-α IL-1β, TNF-α (p < 0.01), and IL-6 (p < 0.05) significantly lower in ketamine group
24 h
Clarke et al. (2017) 16 Adult male CD1 mice LPS (after ketamine) Ketamine 5 or 10 mg/kg i.p. or saline Plasma GM-CSF, IFN-γ, IL-10, IL-1β, IL-6, and TNF-α Both ketamine doses decreased TNF-α (p < 0.01). High-dose ketamine decreased IL-1β (p < 0.05)
2h
Eskelund et al. (2017) 54 FSL rats FSL; genetic rat depression model (A) Ketamine 15 mg/kg i.p. or saline; (B) Ketamine 15 mg/kg i.p. or saline every third day for 14 days Brain and plasma KYN, KynA, QUIN, 3-HK, TRP, and 5-HT No differences
4 h
Ji et al. (2019) ~51 Male Sprague Dawley rats LPS Ketamine 6 mg/kg i.p. or saline Plasma TNF-α, IL-1β, and IL-6. No differences
11.5 h
Li et al. (2019) Male C57BL/6 mice LPS Ketamine 10 mg/kg i.p. or no treatment Hippocampal IL-1β Ketamine decreased IL-1β (p < 0.001)
~3 h
Reus et al. (2017) 8–10 Adult male Wistar rats FST or Open field + splash test (all after ketamine) Ketamine 5 mg/kg i.p. or saline Serum IL-1β, IL-10, and IL-6 No differences
~1.5 h
Reus et al. (2015) 10–12 Juvenile male Wistar rats Maternal deprivation (+ FST on day 13 and 14) Ketamine 15 mg/kg i.p. or saline, once a day for 14 days Serum and CSF IL-1β, IL-6, and TNF-α Ketamine decreased levels of all cytokines; IL-6 in both serum and CSF, IL-1β and TNF-α in serum only (p < 0.05)
~24 h
Tan et al. (2017) 12 Kunming mice Chronic restraint stress 4 h/day on 21 days Ketamine 20 mg/kg i.p. or saline on days 22, 24, and 25 Serum IL-1β, IL-6, and TNF-α Ketamine decreased levels of all cytokines (p < 0.05)
1 h after last infusion
Unal et al. (2015) 12–14 Sprague Dawley rats 4 h restraint stress on 7 days Ketamine 10 mg/kg i.p. or no treatment on day 7 Hippocampal and PFC TNF-α, IL-1β, and IL-6 (mRNA) In hippocampus, ketamine decreased levels of all cytokines (p < 0.05)
4.5 h
Verdonk et al. (2019) 18 Adult male knock-in CX3CR1GFP/+ Mice LPS Ketamine 10 mg/kg, 90 mg/kg, or placebo Brain tissue levels of multiple markers incl. IL-1α, IL-1β, IL-6, IL-10, IFN-γ, TNF-α; and TRP, KYN, KynA, QUIN, 5-HT, and 3-HK Both ketamine doses decreased IL-1α, IL-6, and G-CSF (p < 0.01 for 90 mg/kg, p < 0.05 for 10 mg/kg)
32 h Both ketamine doses decreased QUIN (p < 0.01). 90 mg/kg ketamine reduced 3-HK (p < 0.0001); and increased KynA (p < 0.001), data for 10 mg/kg not given
Walker et al. (2015) Male albino Wistar rats ACTH or saline for 14 days (TR rat model) Ketamine 10 mg/kg i.p. or saline Plasma IL-6, TNF-α, and CRP. TR ketamine responders had higher CRP than TR placebo group (p < 0.05) and non-TR ketamine group (p < 0.001). TR ketamine non-responders had lower TNF-α than TR placebo group (p < 0.05)
~2 h
Walker et al. (2013) Juvenile CD-1 and adult C57BL/6J male mice LPS (after ketamine) Ketamine 6 mg/kg i.p. or saline Plasma IL-1β; plasma and brain IL-6, KYN, and TRP; and brain IDO mRNA No differences
6 h and 28 h
Wang et al. (2015) 40 Adult male Wistar rats. CUMS over 21 days Ketamine 10 mg/kg i.p. (0.5, 1, 2, or 4 h before behavioral tests) or saline (0.5 h before behavioral tests) Hippocampal IL-1β, IL-6, TNF-α, IDO, KYN, and TRP Ketamine decreased IL-1β at 0.5 and 1 h, TNF-α at 2 and 4 h, and IL-6 at all four timepoints (p < 0.05).
1, 1.5, 2.5, or 4.5 h Ketamine decreased KYN/TRP ratio and IDO at all four timepoints (p < 0.05, IDO at 0.5 h p < 0.01)
Xie et al. (2017) 30 Male Sprague Dawley rats Neuropathic pain/SNI Ketamine 20 mg/kg i.p. or saline Serum IL-1β, IL-6, TNF-α In rats with depression-like phenotype (n = 12), Ketamine decreased IL-1β and IL-6 (p < 0.05). Depression phenotype allocated based on behavioral tests
3 days
Yang et al. (2013a) 20 Male Wistar rats FST Ketamine 10 mg/kg i.p. or saline PFC and hippocampal IL-1β and IL-6 Ketamine significantly decreased IL-1β and IL-6 in PFC and hippocampus (p < 0.05)
0.5 h
Yang et al. (2013b) 20 Male Wistar rats LPS Ketamine 10 mg/kg i.p. or saline PFC IL-1β, IL-6, and IL-10 Ketamine significantly decreased IL-1β (p < 0.01) and IL-6 (p < 0.05), and increased IL-10 (p < 0.05; anti-inflammatory)
1 h
Yang et al. (2020) 10 Adult male Sprague Dawley rats Chronic postsurgical pain Ketamine 20 mg/kg i.p. or saline Hippocampal IL-1β, IL-6, and TNF-α Ketamine significantly decreased IL-1β, IL-6 (p < 0.01), and TNF-α (p < 0.001). Only rats showing depression-like phenotype on behavioral tests were investigated
7 days
Zhang et al. (2016) 32 Adult male SD rats Inflammatory pain-induced (CFA) Ketamine 20 mg/kg i.p. or saline Hippocampal IL-6, IL-1β, IDO, KYN, TRP, and 5-HT Ketamine decreased IL-6 at 1 h and 24 h (p < 0.05), and IL-1β at 24 h (p < 0.01). IDO decreased at 1 h and 24 h (p < 0.01), KYN/TRP ratio decreased at 1 h and 24 h (p < 0.05), 5-HT/TRP ratio increased at 1 h and 24 h (p < 0.05)
1 h or 24 h
Zhao et al. (2020) 20 Male Wistar rats LPS Ketamine 10 mg/kg i.p. or no treatment Hippocampal IL-1β, IL-6, TNF-α, and IDO Ketamine decreased IL-6, TNF-α, and IDO (p < 0.01) and IL-1β (p < 0.05)
2 h
Zhu et al. (2015) 12 Adult male Sprague Dawley rats CUMS Ketamine 100 mg/kg i.p. + sham ECT or sham ECT only once daily for 7 days hippocampal IL-1β and TNF-α (mRNA) Ketamine decreased TNF-α (p < 0.05)
9 days

ACTH: adrenocorticotropic hormone; CFA: complete Freund’s adjuvant; CRP: C-reactive protein; CSF: cerebrospinal fluid; CUMS: chronic unpredictable mild stress; ECT: electroconvulsive therapy; FSL: Flinders sensitive line; FST: forced swim test; G-CSF: granulocyte colony-stimulating factor; GM-CSF: granulocyte-macrophage colony-stimulating factor; IDO: indoleamine 2,3-dioxygenase; IL: interleukin; IFN-α: interferon-alpha; i.p.: intraperitoneal; KYN: kynurenine; KynA: kynurenic acid; LPS: lipopolysaccharide; PFC: prefrontal cortex; QUIN: quinolinic acid; SNI: spared nerved injury; TNF-α: tumor necrosis factor-alpha; TRP: tryptophan.

a

Numbers of rats indicate how many rats were included in the biomarker analysis in our groups of interest. If not mentioned otherwise, assumed that all rats in a group analyzed.

b

Outcome measurement timepoints indicate the time from last ketamine administration (or LPS where this is after ketamine) to sample collection. If not mentioned otherwise, assumed that samples taken immediately after behavioral tests.

Quality scoring of animal studies are listed in Supplemental Table S2. Total quality scores ranged between four and eight out of a total of eight. Key differences in study quality arose from the depression model used, specifically whether induced depression was confirmed with behavioral testing ideally comparing performance to a group of nondepressed control rodents. There was also variability in clarity of study protocol and in comprehensiveness of reporting of outcome data and statistics for different comparisons.

Results of individual studies and evidence synthesis

Human studies

Five out of six human studies measuring inflammatory proteins found decreases in at least one marker. Of these, one study stratified patients according to the responder status and found changes only in ketamine responders (Yang et al., 2015b). IL-1β decrease was observed in two out of three studies (p < 0.05; Yang et al., 2015b; Zhan et al., 2020); TNF-α in two out of five studies (p < 0.01; Chen et al., 2018; Zhan et al., 2020); IL-6 in three out of six studies (p < 0.01; Kiraly et al., 2017; Yang et al., 2015b; Zhan et al., 2020); and IFN-γ in one study out of three (p < 0.01; Zhan et al., 2020). Soluble tumor necrosis factor receptor 1 (sTNFR1) was measured in one study and was found reduced (p < 0.01; Park et al., 2017), likewise for IL-23 (Zhan et al., 2020), IL-1α, granulocyte colony-stimulating factor (G-CSF), platelet-derived growth factor (PDGF)-AA (p < 0.05), and interferon gamma-induced protein (IP)-10 (Kiraly et al., 2017). Additionally, IL-8 (p < 0.01), IL-13 (Kiraly et al., 2017), IL-2, IL-7, IL-10, IL-17A, fractalkine (p < 0.01; Zhan et al., 2020), granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-4, IL-5, and IL-12p70 (p < 0.05; Zhan et al., 2020) were each decreased in one out of two studies. Finally, increases in cytokines IL-6 (Park et al., 2017) and IL-7 (Kiraly et al., 2017) were observed in one study each (p < 0.01).

Most changes in markers were short-term, lasting up to 240 min. However, Yang et al. (2015b) observed reductions sustaining for 1 and 3 days for IL-1β and IL-6, respectively, and Kiraly et al. (2017) observed reductions in IL-8 and PDGF-AA and increases in IL-7 at 24 h. Zhan et al. (2020) found no significant changes at the 24-h mark; however, levels of 14 proteins were reduced 2 week post-sixth ketamine infusion. In two of the five studies with significant findings, participants were maintained on their medications (Chen et al., 2018; Zhan et al., 2020), and in one study, BD subjects were allowed mood stabilizers (Park et al., 2017). In the study by Allen et al. (2018), who found no changes in any inflammatory protein, the majority of patients were medicated.

Changes in KYN metabolites indicating decreased inflammation were observed in two studies. Kadriu et al. (2019) found increased KYN, KynA, and KYN/KynA ratio and reduced IDO and QA/KYN ratio. These changes were long-term and were observed 1-day post-infusion for KYN/KynA and QA/KYN ratios, and 1- and 3-day post-infusion for KYN, KynA, and IDO; all p-values <0.01, except from KYN at day 3 (p < 0.05). Zhou et al. (2018) stratified patients according to the responder status and found changes only in ketamine responders; increased KynA and KynA/KYN ratio was measured at 24 h post-first and post-sixth ketamine infusion (all p-values <0.05), with the infusions given over a 12-day period. No changes were observed in TRP.

Yang et al. (2015b) and Moaddel et al. (2018) measured KYN and TRP but found no significant ketamine-induced changes over time. However, Moaddel et al. (2018) observed differential changes in markers when analyzed comparing to a placebo group, specifically a sharp initial drop in KYN and KYN/TRP ratio in the placebo group compared to slight increases with ketamine. Finally, Allen et al. (2018) measured KYN, KynA, and TRP with no significant results, but observed a trend toward decreased KYN at 2 h (p = 0.067) and KYN/TRP ratio at 24 h (p = 0.054) in responders.

Animal studies

About 18 out of 21 animal studies found reductions in one or more pro-inflammatory markers. Decreased IL-1β was observed in 14 out of 19 studies and decreased IL-6 in 13 out of 18 studies. Decreased TNF-α was found in nine out of 14 studies; additionally, a study on adrenocorticotropic hormone (ACTH)-induced treatment resistance by Walker et al. (2015) observed decreased TNF-α in ketamine nonresponders compared to a placebo group, p < 0.05. One study recorded an unexpected surge in TNF-α (Abelaira et al., 2017). IL-1α was measured in one study and found decreased, p < 0.01 (Verdonk et al., 2019). One study out of five also observed an increase in anti-inflammatory cytokine IL-10 (p < 0.05; Yang et al., 2013b). Walker et al. (2015) observed elevated CRP in ACTH-treated ketamine responders compared with placebo group, p < 0.05. IFN-γ was measured twice with no differences found (Clarke et al., 2017; Verdonk et al., 2019).

Four out of six studies measuring TRP metabolites found changes indicating decreased inflammation. Verdonk et al. (2019) observed increased KynA and decreased QUIN and 3-HK in ketamine-treated mice (p < 0.01); however, no changes in these three were observed by Eskelund et al. (2017). KYN/TRP ratio was decreased in two studies out of five (p < 0.05; Wang et al., 2015; Zhang et al., 2016), but no differences were found in absolute levels of KYN or TRP. IDO was decreased in three studies out of four, all p-values <0.01 (Wang et al., 2015; Zhang et al., 2016; Zhao et al., 2020). Two studies found no significant differences in any metabolite of the pathway (Eskelund et al., 2017; Walker et al., 2013).

No remarkable differences appeared between results of studies measuring central versus peripheral markers. Of studies measuring both, Reus et al. (2015) found ketamine to reduce IL-6 in both serum and CSF, but IL-1β and TNF-α in serum only (all p-values <0.05). One study found overall levels of plasma and brain KYN metabolites to be strongly correlated, though main analyses of ketamine’s effect were only done in brain markers (Verdonk et al., 2019). No marked differences appeared between samples obtained at different timepoints.

There was an indication for dose-dependency, with higher doses showing more robust effects. Three out of four studies with a low ketamine dose of 5 or 6 mg/kg found no significant results, with one observing a decrease only in TNF-α but not in IL-1β, which was decreased with a higher 10 mg/kg infusion in the same study (Clarke et al., 2017). One study found reductions in three pro-inflammatory markers after both 10 and 90 mg/kg doses; however, all changes were larger following the higher dose (Verdonk et al., 2019).

Discussion

This study is the first systematic review examining the effect of ketamine on both inflammation and TRP–KYN pathway in depression in clinical as well as preclinical animal studies. Preclinical evidence has brought strong evidence for ketamine’s anti-inflammatory and neuroprotective properties, with nearly all null findings occurring in studies where a low-dose ketamine was used. However, studies in humans are thus far scarce and have yielded more mixed findings, they overall seem to support decreases in inflammation and activation of neuroprotective branch of the KYN pathway, at least in a subset of patients.

Ketamine-induced reductions of inflammatory markers were observed most commonly for the cytokines IL-1β, IL-6, and TNF-α, which have all been consistently found to be implicated in depressive illness (Borsini et al., 2020; Osimo et al., 2020; Raison et al., 2006; Rosenblat et al., 2014). These three cytokines are known to upregulate the enzyme IDO, which converts TRP into KYN (Maes et al., 2007); additionally, IL-1β enhances expression of KMO that further converts KYN into its neurotoxic metabolites (Moffett and Namboodiri, 2003). Besides effect on TRP metabolism, pro-inflammatory cytokines contribute to depressive symptoms by disrupting monoamine metabolism and hypothalamic–pituitary–adrenal (HPA) axis function (Felger and Lotrich, 2013; Nikkheslat et al., 2018, 2020) and also by directly influencing glutamate signaling contributing to glutamate excitotoxicity and reduced BDNF (Haroon and Miller, 2016; Miller et al., 2009). It is notable that in many clinical studies changes in inflammatory proteins did not sustain past 24 h, and it is uncertain whether such transient changes can inflict cascades of downstream events that are implicated in ketamine’s long-term antidepressant effects. Observations of KYN metabolite changes of up to 3 days support this possibility, but further research is called for. The ketamine response has previously been found to be associated with increased BDNF up to 1-week post-infusion (Allen et al., 2015; Haile et al., 2014), yet it is unknown to what extent this is induced by ketamine’s anti-inflammatory effects in contrast to the drug’s direct effect on glutamatergic signaling or other mechanisms.

The IDO was found decreased in all but one study it was measured, supporting ketamine’s anti-inflammatory action through decreasing pro-inflammatory cytokines and subsequently downregulating the activity of the enzyme. In animal models, IDO activity has been found essential for inflammation-induced depressive symptoms (Lawson et al., 2013; O’Connor et al., 2009). In contrast, a large longitudinal study in depressed patients found that KYN/TRP ratio, a commonly used indirect indicator of IDO activity, did not mediate the relationship between inflammation and depressive symptoms; in fact, KYN/TRP ratio was even lower in depressed patients though this was no longer significant after adjusting for antidepressant use (Quak et al., 2014). However, validity of KYN/TRP ratio as a proxy for IDO has been challenged (Badawy and Guillemin, 2019), which may also help explain why reductions in this ratio were observed only in two studies of this review despite more consistently observed changes in IDO. In future investigations, direct measurement of IDO is endorsed for reliable evidence of the enzyme’s activity.

Although decreased IDO may lead to reduced synthesis of KYN, our review found no consistent evidence for changes in this metabolite. Changes in KYN may be less detectable due to its eventual transamination into downstream metabolites (Badawy and Guillemin, 2019), levels of which are arguably more relevant markers for ketamine’s action and anti-inflammatory effects. Indeed, as an indicator of inflammation, increased IDO only leads to neurotoxicity with additional upregulation of KMO and subsequent conversion of KYN into its neurotoxic metabolites. Our review found evidence for increased KynA and reduced QUIN following ketamine in both clinical and preclinical studies, alongside decreased 3-HK in one animal study, supporting anti-inflammatory activity and activation of KAT over KMO and a shift toward neuroprotective rather than neurotoxic pathway. The effect of ketamine in decreasing pro-inflammatory cytokines leads to less activity of KMO and increased availability and synthesis of KYN into KynA instead of QUIN. KMO has been found essential for inflammation-induced depression in rodents (Parrott et al., 2016), suggesting the present results could be of high clinical relevance. However, due to the small number of studies measuring these metabolites as presented in our review, further research is needed.

Heterogeneity in inclusion criteria and methodologies of included human studies not only complicate the interpretation of the evidence base but also shed light on the potential reasons for the inconsistencies seen in the literature. Studies without significant findings appeared either underpowered and were on medicated patients (Allen et al., 2018) or measured only a few markers and applied less strict treatment-resistant criteria (Moaddel et al., 2018). Given inflammation levels are generally lower in nontreatment-resistant patients (Cattaneo et al., 2020; Strawbridge et al., 2015; Yang et al., 2019),; and traditional antidepressants (Köhler et al., 2018; Strawbridge et al., 2015) and mood stabilizers (Li et al., 2015; Rapaport et al., 1999) have been found to exert anti-inflammatory effects, ketamine might be less likely to show anti-inflammatory effects in nontreatment-resistant and in medicated subjects. It is yet worth noting that even in many higher-quality studies decreases were observed only in one or two inflammatory markers. The two studies that stratified results according to the responder status to antidepressants only found significant results in those who responded, supporting the possibility that ketamine’s antidepressant effect might at least partially be mediated through its anti-inflammatory effects, which are more prominent in responders (Yang et al., 2015b; Zhou et al., 2018).

Of relevance, surgical and preclinical research has indicated ketamine does not reduce inflammation unless it is abnormally high (Loix et al., 2011); whether and to what extent baseline inflammation predicts ketamine’s effect on inflammatory markers as well as symptomatic improvement of depression are important areas for further investigation. In the study by Yang et al. (2015b), who found both higher baseline levels of, as well as ketamine-induced reductions in, IL-6 and IL-1β to be associated with response status, baseline levels of these markers were remarkably higher than seen in most populations. Variability in baseline inflammation levels within and between studies could also be one reason why preclinical findings have not been replicated as consistently in humans; in rodents, depression is induced mechanistically and in many times with inflammatory stimuli, providing optimal condition for ketamine to demonstrate its anti-pro-inflammatory effects. The only animal study with a moderate-to-high ketamine dose but with no significant findings used a genetic rat model of depression (Eskelund et al., 2017), which had previously demonstrated lower levels of QUIN and no difference in KYN/TRP levels compared to its selectively bred controls (Eskelund et al., 2016). It is also notable that in several rodent studies ketamine was administered before inflammatory challenge, which can result in more potent anti-inflammatory effects than when administered once inflammation is already present (Taniguchi et al., 2001).

There was a strong indication for dose-dependency across animal studies, and likewise one clinical trial found anti-inflammatory effects only following 0.5 mg/kg but not 0.2 mg/kg ketamine infusion (Chen et al., 2018). Potential increased efficacy of even higher ketamine doses in humans is unclear. In surgical contexts, subanesthetic doses similar or even lower to antidepressant doses have been found sufficient for inducing anti-inflammatory effects (Dale et al., 2012). Evidence of antidepressant efficacy of higher ketamine doses is mixed with one study showing no superiority of 1.0 mg/kg over 0.5 mg/kg infusion (Fava et al., 2018), but another indicating more pronounced responses after doses were escalated from 0.5 mg/kg to 0.75 mg/kg (Cusin et al., 2017). Hypothetically, it remains plausible higher ketamine dose could also trigger or augment anti-inflammatory effect in some patients in whom this was not evident and could therefore be investigated in the future. Research on the effect of multiple ketamine infusions is also crucial, given repeated dosing is commonly needed and used in practice (Phillips et al., 2019; Voort et al., 2016).

It is important to highlight that the included human studies only measured circulating markers, which might not provide a reflection of levels in the central nervous system. Regarding TRP metabolites, KYN, TRP, and 3-HK readily cross the blood–brain barrier, but QUIN and KynA do this at lower rates and are instead produced in the brain immune cells (Guillemin, 2012; Schwarcz and Pellicciari, 2002). Although inflammation has previously been suggested to increase blood–brain barrier permeability (Skaper, 2017), this notion has recently been challenged (Turkheimer et al., 2020) and robust consistent associations between brain and blood inflammation, or KYNs are yet to be found in depression (Nettis and Pariante, 2020). A recent study in unmedicated-depressed patients showed that while plasma and CSF levels of KYN, KYN/TRP ratio, and QUIN correlated strongly, there was no significant relationship for KynA, TRP, and QUIN/KynA ratio (Haroon et al., 2020). In the same study, a significant relationship was found between plasma and CSF CRP and IL-6 soluble receptor, but correlations were close to zero for most other inflammatory proteins including TNF, IL-6, and IL-1β (Haroon et al., 2020). Studies examining markers in the CSF or in the brain with positron emission tomography are therefore urgently needed.

There are some limitations with the current review that may help for directions of future studies. The number of clinical studies on the effect of ketamine is still small, and relying only on using animals to model complex psychiatric conditions and investigate the effectiveness of drugs would not be an ideal approach (O’Leary and Cryan, 2013). Gray literature was not searched in this review. Meta-analysis could not be conducted due to variability between methodologies and biomarkers measured of the included studies. Heterogeneity between studies, namely in diagnoses, disease profile, potential comorbidities, medication status, and analysis methods, highlights the need for research with more comparable inclusion criteria and methodology. Further, IFN-γ was only measured three times and CRP twice; IFN-γ is the key cytokine activating IDO (Maes et al., 2007); and CRP has been found as a predictor for response to antidepressants with anti-inflammatory characteristics (Yang et al., 2019); therefore, inclusion of these markers in future research would be of interest. Future studies should also ideally include a placebo group, highlighted by saline-induced fluctuations in markers observed in some studies (Chen et al., 2018; Moaddel et al., 2018).

Conclusion

In conclusion, the present review supports ketamine’s anti-inflammatory effects in depressed humans and rodents. Ketamine is considered the most effective antidepressant available for treatment-resistant patients and for suicide prevention, enhancing understanding of its pharmacology is crucial for the development of precision medicine, understanding of neurobiological mechanisms underlying depression, and identification of therapeutic targets for other novel antidepressants with potentially better side effect profile and less abuse liability. Crucial next steps for further research include investigation of the specific molecular mechanism behind ketamine’s immunomodulatory effects, examination of clinical relevance of inflammatory changes, and measurement of markers in the central nervous system.

Supplemental Material

sj-docx-1-jop-10.1177_02698811211026426 – Supplemental material for Ketamine’s effect on inflammation and kynurenine pathway in depression: A systematic review

Supplemental material, sj-docx-1-jop-10.1177_02698811211026426 for Ketamine’s effect on inflammation and kynurenine pathway in depression: A systematic review by Emma Kopra, Valeria Mondelli, Carmine Pariante and Naghmeh Nikkheslat in Journal of Psychopharmacology

Footnotes

Declaration of conflicting interests: The author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: Dr Valeria Mondelli is supported by MQ: Transforming Mental Health (Grant: MQBF1) and by the Medical Research Foundation (Grant number MRF-160-0005-ELP-MONDE). Prof Carmine Pariante has received research funding from the Medical Research Council (UK) and the Wellcome Trust for research on depression and inflammation as part of two large consortia that also include Johnson & Johnson, GSK, and Lundbeck.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This systematic review is prepared as part of a clinical study to investigate the anti-inflammatory effect of ketamine funded by the NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London to Dr Naghmeh Nikkheslat. The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR, or the Department of Health.

Supplemental material: Supplemental material for this article is available online.

References

  1. Abelaira HM, Reus GZ, Ignacio ZM, et al. (2017) Ketamine exhibits different neuroanatomical profile after mammalian target of rapamycin inhibition in the prefrontal cortex: The role of inflammation and oxidative stress. Mol Neurobiol 54: 5335–5346. [DOI] [PubMed] [Google Scholar]
  2. Allen AP, Naughton M, Dowling J, et al. (2015) Serum BDNF as a peripheral biomarker of treatment-resistant depression and the rapid antidepressant response: A comparison of ketamine and ECT. J Affect Disord 186: 306–311. [DOI] [PubMed] [Google Scholar]
  3. Allen AP, Naughton M, Dowling J, et al. (2018) Kynurenine pathway metabolism and the neurobiology of treatment-resistant depression: Comparison of multiple ketamine infusions and electroconvulsive therapy. J Psychiatr Res 100: 24–32. [DOI] [PubMed] [Google Scholar]
  4. Andersson NW, Gustafsson LN, Okkels N, et al. (2015) Depression and the risk of autoimmune disease: A nationally representative, prospective longitudinal study. Psychol Med 45: 3559. [DOI] [PubMed] [Google Scholar]
  5. Aricioglu F, Yalcinkaya C, Ozkartal CS, et al. (2020) NLRP1-mediated antidepressant effect of ketamine in chronic unpredictable mild stress model in rats. Psychiatry Investig 17: 283–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Badawy AA, Guillemin G. (2019) The plasma [Kynurenine]/[Tryptophan] ratio and indoleamine 2, 3-dioxygenase: Time for appraisal. Int J Tryptophan Res 12: 1178646919868978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Benros ME, Waltoft BL, Nordentoft M, et al. (2013) Autoimmune diseases and severe infections as risk factors for mood disorders: A nationwide study. JAMA Psychiatry 70: 812–820. [DOI] [PubMed] [Google Scholar]
  8. Berman RM, Cappiello A, Anand A, et al. (2000) Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 47: 351–354. [DOI] [PubMed] [Google Scholar]
  9. Bobo WV, Vande Voort JL, Croarkin PE, et al. (2016) Ketamine for treatment-resistant unipolar and bipolar major depression: Critical review and implications for clinical practice. Depress Anxiety 33: 698–710. [DOI] [PubMed] [Google Scholar]
  10. Borsini A, Di Benedetto MG, Giacobbe J, et al. (2020) Pro-and anti-inflammatory properties of interleukin (IL6) in vitro: Relevance for major depression and for human hippocampal neurogenesis. Int J Neuropsychopharmacol 23: 738–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bryleva EY, Brundin L. (2017) Kynurenine pathway metabolites and suicidality. Neuropharmacology 112: 324–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Cattaneo A, Ferrari C, Turner L, et al. (2020) Whole blood expression of inflammasome-and glucocorticoid-related mRNAs correctly separates treatment-resistant depressed patients from drug-free responsive patients in the BIODEP study. Transl Psychiatry 10: 352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Cattaneo A, Ferrari C, Uher R, et al. (2016) Absolute measurements of macrophage migration inhibitory factor and interleukin-1-β mRNA levels accurately predict treatment response in depressed patients. Int J Neuropsychopharmacol 19: pyw045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cattaneo A, Gennarelli M, Uher R, et al. (2013) Candidate genes expression profile associated with antidepressants response in the GENDEP study: Differentiating between baseline ‘predictors’ and longitudinal ‘targets’. Neuropsychopharmacology 38: 377–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Chang D, Zhao J, Zhang X, et al. (2019) Effect of ketamine combined with DHA on lipopolysaccharide-induced depression-like behavior in rats. Int Immunopharmacol 75: 105788. [DOI] [PubMed] [Google Scholar]
  16. Chen MH, Li CT, Lin WC, et al. (2018) Rapid inflammation modulation and antidepressant efficacy of a low-dose ketamine infusion in treatment-resistant depression: A randomized, double-blind control study. Psychiatry Res 269: 207–211. [DOI] [PubMed] [Google Scholar]
  17. Clarke M, Razmjou S, Prowse N, et al. (2017) Ketamine modulates hippocampal neurogenesis and pro-inflammatory cytokines but not stressor induced neurochemical changes. Neuropharmacology 112: 210–220. [DOI] [PubMed] [Google Scholar]
  18. Colín-González AL, Maldonado PD, Santamaría A. (2013) 3-Hydroxykynurenine: An intriguing molecule exerting dual actions in the central nervous system. Neurotoxicology 34: 189–204. [DOI] [PubMed] [Google Scholar]
  19. Corriger A, Pickering G. (2019) Ketamine and depression: A narrative review. Drug Des Devel Ther 13: 3051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Coyle CM, Laws KR. (2015) The use of ketamine as an antidepressant: A systematic review and meta-analysis. Hum Psychopharmacol 30: 152–163. [DOI] [PubMed] [Google Scholar]
  21. Cusin C, Ionescu DF, Pavone KJ, et al. (2017) Ketamine augmentation for outpatients with treatment-resistant depression: Preliminary evidence for two-step intravenous dose escalation. Aust N Z J Psychiatry 51: 55–64. [DOI] [PubMed] [Google Scholar]
  22. Dale O, Somogyi AA, Li Y, et al. (2012) Does intraoperative ketamine attenuate inflammatory reactivity following surgery? A systematic review and meta-analysis. Anesth Analg 115: 934–943. [DOI] [PubMed] [Google Scholar]
  23. De Kock M, Loix S, Lavand’homme P. (2013) Ketamine and peripheral inflammation. CNS Neurosci Ther 19: 403–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Diazgranados N, Ibrahim L, Brutsche NE, et al. (2010) A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-resistant bipolar depression. Arch Gen Psychiatry 67: 793–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Duman RS, Aghajanian GK. (2012) Synaptic dysfunction in depression: Potential therapeutic targets. Science 338: 68–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Enache D, Pariante CM, Mondelli V. (2019) Markers of central inflammation in major depressive disorder: A systematic review and meta-analysis of studies examining cerebrospinal fluid, positron emission tomography and post-mortem brain tissue. Brain Behav Immun 81: 24–40. [DOI] [PubMed] [Google Scholar]
  27. Eskelund A, Budac DP, Sanchez C, et al. (2016) Female Flinders Sensitive Line rats show estrous cycle-independent depression-like behavior and altered tryptophan metabolism. Neuroscience 329: 337–348. [DOI] [PubMed] [Google Scholar]
  28. Eskelund A, Li Y, Budac DP, et al. (2017) Drugs with antidepressant properties affect tryptophan metabolites differently in rodent models with depression-like behavior. J Neurochem 142: 118–131. [DOI] [PubMed] [Google Scholar]
  29. Fava M, Freeman MP, Flynn M, et al. (2018) Double-blind, placebo-controlled, dose-ranging trial of intravenous ketamine as adjunctive therapy in treatment-resistant depression (TRD). Mol Psychiatry 25: 1592–1603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. FDA (2019) FDA approves new nasal spray medication for treatment-resistant depression; available only at a certified doctor’s office or clinic. Available at: https://www.fda.gov/news-events/press-announcements/fda-approves-new-nasal-spray-medication-treatment-resistant-depression-available-only-certified (accessed 1 October 2020).
  31. Felger JC, Lotrich FE. (2013) Inflammatory cytokines in depression: Neurobiological mechanisms and therapeutic implications. Neuroscience 246: 199–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Ferrari AJ, Charlson FJ, Norman RE, et al. (2013) Burden of depressive disorders by country, sex, age, and year: Findings from the global burden of disease study 2010. PLoS Med 10: e1001547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Gabbay V, Klein RG, Katz Y, et al. (2010) The possible role of the kynurenine pathway in adolescent depression with melancholic features. J Child Psychol Psychiatry 51: 935–943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Ganong AH, Cotman CW. (1986) Kynurenic acid and quinolinic acid act at N-methyl-D-aspartate receptors in the rat hippocampus. J Pharmacol Exp Ther 236: 293–299. [PubMed] [Google Scholar]
  35. Gaynes BN, Warden D, Trivedi MH, et al. (2009) What did STAR* D teach us? Results from a large-scale, practical, clinical trial for patients with depression. Psychiatr Serv 60: 1439–1445. [DOI] [PubMed] [Google Scholar]
  36. Guillemin GJ. (2012) Quinolinic acid, the inescapable neurotoxin. FEBS J 279: 1356–1365. [DOI] [PubMed] [Google Scholar]
  37. Haile CN, Murrough JW, Iosifescu DV, et al. (2014) Plasma brain derived neurotrophic factor (BDNF) and response to ketamine in treatment-resistant depression. Int J Neuropsychopharmacol 17: 331–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Haroon E, Miller AH. (2016) Inflammation effects on brain glutamate in depression: Mechanistic considerations and treatment implications. In: Dantzer R, Capuron L. (eds) Inflammation-Associated Depression: Evidence, Mechanisms and Implications. Cham: Springer, pp.173–198. [DOI] [PubMed] [Google Scholar]
  39. Haroon E, Welle JR, Woolwine BJ, et al. (2020) Associations among peripheral and central kynurenine pathway metabolites and inflammation in depression. Neuropsychopharmacology 45: 998–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Hashimoto K. (2019) Rapid-acting antidepressant ketamine, its metabolites and other candidates: A historical overview and future perspective. Psychiatry Clin Neurosci 73: 613–627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Hestad KA, Engedal K, Whist JE, et al. (2017) The relationships among tryptophan, kynurenine, indoleamine 2, 3-dioxygenase, depression, and neuropsychological performance. Front Psychol 8: 1561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Hughes MM, Carballedo A, McLoughlin DM, et al. (2012) Tryptophan depletion in depressed patients occurs independent of kynurenine pathway activation. Brain Behav Immun 26: 979–987. [DOI] [PubMed] [Google Scholar]
  43. Jelen LA, Young AH, Stone JM. (2020) Ketamine: A tale of two enantiomers. J Psychopharmacol 35: 109–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Ji M, Mao M, Li S, et al. (2019) Acute ketamine administration attenuates lipopolysaccharide-induced depressive-like behavior by reversing abnormal regional homogeneity in the nucleus accumbens. Neuroreport 30: 421–427. [DOI] [PubMed] [Google Scholar]
  45. Kadriu B, Farmer CA, Yuan P, et al. (2019) The kynurenine pathway and bipolar disorder: Intersection of the monoaminergic and glutamatergic systems and immune response. Mol Psychiatry. Epub ahead of print 15 November 2019. DOI: 10.1038/s41380-019-0589-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Khandaker GM, Pearson RM, Zammit S, et al. (2014) Association of serum interleukin 6 and C-reactive protein in childhood with depression and psychosis in young adult life: A population-based longitudinal study. JAMA Psychiatry 71: 1121–1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Kiraly DD, Horn SR, Van Dam NT, et al. (2017) Altered peripheral immune profiles in treatment-resistant depression: Response to ketamine and prediction of treatment outcome. Transl Psychiatry 7: e1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Kishimoto T, Chawla JM, Hagi K, et al. (2016) Single-dose infusion ketamine and non-ketamine N-methyl-d-aspartate receptor antagonists for unipolar and bipolar depression: A meta-analysis of efficacy, safety and time trajectories. Psychol Med 46: 1459–1472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Köhler CA, Freitas TH, Stubbs B, et al. (2018) Peripheral alterations in cytokine and chemokine levels after antidepressant drug treatment for major depressive disorder: Systematic review and meta-analysis. Mol Neurobiol 55: 4195–4206. [DOI] [PubMed] [Google Scholar]
  50. Lanquillon S, Krieg J, Bening-Abu-Shach U, et al. (2000) Cytokine production and treatment response in major depressive disorder. Neuropsychopharmacology 22: 370–379. [DOI] [PubMed] [Google Scholar]
  51. Larkin GL, Beautrais AL. (2011) A preliminary naturalistic study of low-dose ketamine for depression and suicide ideation in the emergency department. Int J Neuropsychopharmacol 14: 1127–1131. [DOI] [PubMed] [Google Scholar]
  52. Lawson MA, Parrott JM, McCusker RH, et al. (2013) Intracerebroventricular administration of lipopolysaccharide induces indoleamine-2, 3-dioxygenase-dependent depression-like behaviors. J Neuroinflammation 10: 875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Li H, Hong W, Zhang C, et al. (2015) IL-23 and TGF-β1 levels as potential predictive biomarkers in treatment of bipolar I disorder with acute manic episode. J Affect Disord 174: 361–366. [DOI] [PubMed] [Google Scholar]
  54. Li JM, Liu LL, Su WJ, et al. (2019) Ketamine may exert antidepressant effects via suppressing NLRP3 inflammasome to upregulate AMPA receptors. Neuropharmacology 146: 149–153. [DOI] [PubMed] [Google Scholar]
  55. Loix S, De Kock M, Henin P. (2011) The anti-inflammatory effects of ketamine: State of the art. Acta Anaesthesiol Belg 62: 47–58. [PubMed] [Google Scholar]
  56. Lugo-Huitrón R, Ugalde Muñiz P, Pineda B, et al. (2013) Quinolinic acid: An endogenous neurotoxin with multiple targets. Oxid Med Cell Longev 2013: 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Maes M, Mihaylova I, Ruyter MD, et al. (2007) The immune effects of TRYCATs (tryptophan catabolites along the IDO pathway): Relevance for depression–And other conditions characterized by tryptophan depletion induced by inflammation. Neuro Endocrinol Lett 28: 826–831. [PubMed] [Google Scholar]
  58. Miller AH, Maletic V, Raison CL. (2009) Inflammation and its discontents: The role of cytokines in the pathophysiology of major depression. Biol Psychiatry 65: 732–741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Miller CL, Llenos IC, Cwik M, et al. (2008) Alterations in kynurenine precursor and product levels in schizophrenia and bipolar disorder. Neurochem Int 52: 1297–1303. [DOI] [PubMed] [Google Scholar]
  60. Moaddel R, Shardell M, Khadeer M, et al. (2018) Plasma metabolomic profiling of a ketamine and placebo crossover trial of major depressive disorder and healthy control subjects. Psychopharmacology 235: 3017–3030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Moffett JR, Namboodiri MA. (2003) Tryptophan and the immune response. Immunol Cell Biol 81: 247–265. [DOI] [PubMed] [Google Scholar]
  62. Murrough JW, Iosifescu DV, Chang LC, et al. (2013. a) Antidepressant efficacy of ketamine in treatment-resistant major depression: A two-site randomized controlled trial. Am J Psychiatry 170: 1134–1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Murrough JW, Perez AM, Pillemer S, et al. (2013. b) Rapid and longer-term antidepressant effects of repeated ketamine infusions in treatment-resistant major depression. Biol Psychiatry 74: 250–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Nemeroff CB. (2007) Prevalence and management of treatment-resistant depression. J Clin Psychiatry 68: 17. [PubMed] [Google Scholar]
  65. Nettis MA, Pariante CM. (2020) Is there neuroinflammation in depression? Understanding the link between the brain and the peripheral immune system in depression. In: Clow A and Smyth N (eds) Stress and Brain Health: In Clinical Conditions. International Review of Neurobiology. Cambridge, MA: Elsevier, pp.23–40. [DOI] [PubMed] [Google Scholar]
  66. Newport DJ, Carpenter LL, McDonald WM, et al. (2015) Ketamine and other NMDA antagonists: Early clinical trials and possible mechanisms in depression. Am J Psychiatry 172: 950–966. [DOI] [PubMed] [Google Scholar]
  67. Nikkheslat N, McLaughlin AP, Hastings C, et al. (2020) Childhood trauma, HPA axis activity and antidepressant response in patients with depression. Brain Behav Immun 87: 229–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Nikkheslat N, Pariante CM, Zunszain PA. (2018) Neuroendocrine abnormalities in major depression: An insight into glucocorticoids, cytokines, and the kynurenine pathway. In: Baune BT. (ed.) Inflammation and Immunity in Depression. London; San Diego, CA: Elsevier, pp.45–60. [Google Scholar]
  69. O’Connor JC, Lawson MA, Andre C, et al. (2009) Lipopolysaccharide-induced depressive-like behavior is mediated by indoleamine 2, 3-dioxygenase activation in mice. Mol Psychiatry 14: 511–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Ogyu K, Kubo K, Noda Y, et al. (2018) Kynurenine pathway in depression: A systematic review and meta-analysis. Neurosci Biobehav Rev 90: 16–25. [DOI] [PubMed] [Google Scholar]
  71. O’Leary OF, Cryan JF. (2013) Towards translational rodent models of depression. Cell Tissue Res 354: 141–153. [DOI] [PubMed] [Google Scholar]
  72. Osimo EF, Pillinger T, Rodriguez IM, et al. (2020) Inflammatory markers in depression: A meta-analysis of mean differences and variability in 5,166 patients and 5,083 controls. Brain Behav Immun 87: 901–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Pacchiarotti I, Bond DJ, Baldessarini RJ, et al. (2013) The International Society for Bipolar Disorders (ISBD) task force report on antidepressant use in bipolar disorders. Am J Psychiatry 170: 1249–1262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Park M, Newman LE, Gold PW, et al. (2017) Change in cytokine levels is not associated with rapid antidepressant response to ketamine in treatment-resistant depression. J Psychiatr Res 84: 113–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Parrott JM, Redus L, Santana-Coelho D, et al. (2016) Neurotoxic kynurenine metabolism is increased in the dorsal hippocampus and drives distinct depressive behaviors during inflammation. Transl Psychiatry 6: e918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Phillips JL, Norris S, Talbot J, et al. (2019) Single, repeated, and maintenance ketamine infusions for treatment-resistant depression: A randomized controlled trial. Am J Psychiatry 176: 401–409. [DOI] [PubMed] [Google Scholar]
  77. Potter MC, Elmer GI, Bergeron R, et al. (2010) Reduction of endogenous kynurenic acid formation enhances extracellular glutamate, hippocampal plasticity, and cognitive behavior. Neuropsychopharmacology 35: 1734–1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Quak J, Doornbos B, Roest AM, et al. (2014) Does tryptophan degradation along the kynurenine pathway mediate the association between pro-inflammatory immune activity and depressive symptoms? Psychoneuroendocrinology 45: 202–210. [DOI] [PubMed] [Google Scholar]
  79. Raison CL, Capuron L, Miller AH. (2006) Cytokines sing the blues: Inflammation and the pathogenesis of depression. Trends Immunol 27: 24–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Raison CL, Dantzer R, Kelley KW, et al. (2010) CSF concentrations of brain tryptophan and kynurenines during immune stimulation with IFN-α: Relationship to CNS immune responses and depression. Mol Psychiatry 15: 393–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Rapaport MH, Guylai L, Whybrow P. (1999) Immune parameters in rapid cycling bipolar patients before and after lithium treatment. J Psychiatr Res 33: 335–340. [DOI] [PubMed] [Google Scholar]
  82. Reus GZ, Matias BI, Maciel AL, et al. (2017) Mechanism of synergistic action on behavior, oxidative stress and inflammation following co-treatment with ketamine and different antidepressant classes. Pharmacol Rep 69: 1094–1102. [DOI] [PubMed] [Google Scholar]
  83. Reus GZ, Nacif MP, Abelaira HM, et al. (2015) Ketamine ameliorates depressive-like behaviors and immune alterations in adult rats following maternal deprivation. Neurosci Lett 584: 83–87. [DOI] [PubMed] [Google Scholar]
  84. Rosenblat JD, Cha DS, Mansur RB, et al. (2014) Inflamed moods: A review of the interactions between inflammation and mood disorders. Prog Neuropsychopharmacol Biol Psychiatry 53: 23–34. [DOI] [PubMed] [Google Scholar]
  85. Rosenblat JD, McIntyre RS. (2017) Bipolar disorder and immune dysfunction: Epidemiological findings, proposed pathophysiology and clinical implications. Brain Sci 7: 144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Schwarcz R, Pellicciari R. (2002) Manipulation of brain kynurenines: Glial targets, neuronal effects, and clinical opportunities. J Pharmacol Exp Ther 303: 1–10. [DOI] [PubMed] [Google Scholar]
  87. Sforzini L, Pariante CM, Palacios JE, et al. (2019) Inflammation associated with coronary heart disease predicts onset of depression in a three-year prospective follow-up: A preliminary study. Brain Behav Immun 81: 659–664. [DOI] [PubMed] [Google Scholar]
  88. Short B, Fong J, Galvez V, et al. (2018) Side-effects associated with ketamine use in depression: A systematic review. Lancet Psychiatry 5: 65–78. [DOI] [PubMed] [Google Scholar]
  89. Skaper SD. (2017) Impact of inflammation on the blood–neural barrier and blood–nerve interface: From review to therapeutic preview. In: Sharma HS and Sharma A (eds) Nanomedicine in Central Nervous System Injury and Repair. International Review of Neurobiology. Cambridge, MA; San Diego, CA; Oxford; London: Elsevier, pp.29–45. [DOI] [PubMed] [Google Scholar]
  90. Strawbridge R, Arnone D, Danese A, et al. (2015) Inflammation and clinical response to treatment in depression: A meta-analysis. Eur Neuropsychopharmacol 25: 1532–1543. [DOI] [PubMed] [Google Scholar]
  91. Tan S, Wang Y, Chen K, et al. (2017) Ketamine alleviates depressive-like behaviors via down-regulating inflammatory cytokines induced by chronic restraint stress in mice. Biol Pharm Bull 40: 1260–1267. [DOI] [PubMed] [Google Scholar]
  92. Taniguchi T, Shibata K, Yamamoto K. (2001) Ketamine inhibits endotoxin-induced shock in rats. Anesthesiology 95: 928–932. [DOI] [PubMed] [Google Scholar]
  93. Turkheimer FE, Althubaity N, Schubert J, et al. (2020) Increased serum peripheral C-reactive protein is associated with reduced brain barriers permeability of TSPO radioligands in healthy volunteers and depressed patients: Implications for inflammation and depression. Brain Behav Immun 91: 487–497. [DOI] [PubMed] [Google Scholar]
  94. Unal G, Terzioglu G, Sahin C, et al. (2015) Acute administration of ketamine inhibited stress induced pro-inflammatory cytokines in hippocampus of rats. Klinik Psikofarmakol Bulteni 25(Suppl. 1): S101. [Google Scholar]
  95. Valkanova V, Ebmeier KP, Allan CL. (2013) CRP, IL-6 and depression: a systematic review and meta-analysis of longitudinal studies. J Affect Disord 150: 736–744. [DOI] [PubMed] [Google Scholar]
  96. Verdonk F, Petit AC, AbdelAhad P, et al. (2019) Microglial production of quinolinic acid as a target and a biomarker of the antidepressant effect of ketamine. Brain Behav Immun 81: 361–373. [DOI] [PubMed] [Google Scholar]
  97. Vieta E, Locklear J, Günther O, et al. (2010) Treatment options for bipolar depression: A systematic review of randomized, controlled trials. J Clin Psychopharmacol 30: 579–590. [DOI] [PubMed] [Google Scholar]
  98. Voort JLV, Morgan RJ, Kung S, et al. (2016) Continuation phase intravenous ketamine in adults with treatment-resistant depression. J Affect Disord 206: 300–304. [DOI] [PubMed] [Google Scholar]
  99. Walker AJ, Foley BM, Sutor SL, et al. (2015) Peripheral proinflammatory markers associated with ketamine response in a preclinical model of antidepressant-resistance. Behav Brain Res 293: 198–202. [DOI] [PubMed] [Google Scholar]
  100. Walker AK, Budac DP, Bisulco S, et al. (2013) NMDA receptor blockade by ketamine abrogates lipopolysaccharide-induced depressive-like behavior in C57BL/6J mice. Neuropsychopharmacology 38: 1609–1616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Wang L, Chiang J, Chen S, et al. (2018) Systemic autoimmune diseases are associated with an increased risk of bipolar disorder: A nationwide population-based cohort study. J Affect Disord 227: 31–37. [DOI] [PubMed] [Google Scholar]
  102. Wang N, Yu HY, Shen XF, et al. (2015) The rapid antidepressant effect of ketamine in rats is associated with down-regulation of pro-inflammatory cytokines in the hippocampus. Ups J Med Sci 120: 241–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Wilkinson ST, Ballard ED, Bloch MH, et al. (2018) The effect of a single dose of intravenous ketamine on suicidal ideation: A systematic review and individual participant data meta-analysis. Am J Psychiatry 175: 150–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Xie ZM, Wang XM, Xu N, et al. (2017) Alterations in the inflammatory cytokines and brain-derived neurotrophic factor contribute to depression-like phenotype after spared nerve injury: Improvement by ketamine. Sci Rep 7: 3124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Yang C, Hong T, Shen J, et al. (2013. a) Ketamine exerts antidepressant effects and reduces IL-1beta and IL-6 levels in rat prefrontal cortex and hippocampus. Exp Ther Med 5: 1093–1096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Yang C, Shen J, Hong T, et al. (2013. b) Effects of ketamine on lipopolysaccharide-induced depressive-like behavior and the expression of inflammatory cytokines in the rat prefrontal cortex. Mol Med Rep 8: 887–890. [DOI] [PubMed] [Google Scholar]
  107. Yang C, Shirayama Y, Zhang JC, et al. (2015. a) R-ketamine: A rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry 5: e632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Yang C, Wardenaar KJ, Bosker FJ, et al. (2019) Inflammatory markers and treatment outcome in treatment resistant depression: A systematic review. J Affect Disord 257: 640–649. [DOI] [PubMed] [Google Scholar]
  109. Yang J, Wang N, Yang C, et al. (2015. b) Serum interleukin-6 is a predictive biomarker for ketamine’s antidepressant effect in treatment-resistant patients with major depression. Biol Psychiatry 77: e19–e20. [DOI] [PubMed] [Google Scholar]
  110. Yang Y, Song Y, Zhang X, et al. (2020) Ketamine relieves depression-like behaviors induced by chronic postsurgical pain in rats through anti-inflammatory, anti-oxidant effects and regulating BDNF expression. Psychopharmacology 237: 1657–1669. [DOI] [PubMed] [Google Scholar]
  111. Zanos P, Moaddel R, Morris PJ, et al. (2016) NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature 533: 481–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Zhan Y, Zhou Y, Zheng W, et al. (2020) Alterations of multiple peripheral inflammatory cytokine levels after repeated ketamine infusions in major depressive disorder. Transl Psychiatry 10: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Zhang GF, Wang J, Han JF, et al. (2016) Acute single dose of ketamine relieves mechanical allodynia and consequent depression-like behaviors in a rat model. Neurosci Lett 631: 7–12. [DOI] [PubMed] [Google Scholar]
  114. Zhao J, Liu X, Chang D, et al. (2020) Low-dose ketamine improves LPS-induced depression-like behavior in rats by activating cholinergic anti-inflammatory pathways. ACS Chem Neurosci 11: 752–762. [DOI] [PubMed] [Google Scholar]
  115. Zheng W, Zhou Y, Liu W, et al. (2018) Rapid and longer-term antidepressant effects of repeated-dose intravenous ketamine for patients with unipolar and bipolar depression. J Psychiatr Res 106: 61–68. [DOI] [PubMed] [Google Scholar]
  116. Zhou Y, Zheng W, Liu W, et al. (2018) Antidepressant effect of repeated ketamine administration on kynurenine pathway metabolites in patients with unipolar and bipolar depression. Brain Behav Immun 74: 205–212. [DOI] [PubMed] [Google Scholar]
  117. Zhu X, Li P, Hao X, et al. (2015) Ketamine-mediated alleviation of electroconvulsive shock-induced memory impairment is associated with the regulation of neuroinflammation and soluble amyloid-beta peptide in depressive-like rats. Neurosci Lett 599: 32–37. [DOI] [PubMed] [Google Scholar]
  118. Zunszain PA, Anacker C, Cattaneo A, et al. (2012) Interleukin-1β: A new regulator of the kynurenine pathway affecting human hippocampal neurogenesis. Neuropsychopharmacology 37: 939–949. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

sj-docx-1-jop-10.1177_02698811211026426 – Supplemental material for Ketamine’s effect on inflammation and kynurenine pathway in depression: A systematic review

Supplemental material, sj-docx-1-jop-10.1177_02698811211026426 for Ketamine’s effect on inflammation and kynurenine pathway in depression: A systematic review by Emma Kopra, Valeria Mondelli, Carmine Pariante and Naghmeh Nikkheslat in Journal of Psychopharmacology


Articles from Journal of Psychopharmacology (Oxford, England) are provided here courtesy of SAGE Publications

RESOURCES