Hostname: page-component-78c5997874-dh8gc Total loading time: 0 Render date: 2024-11-10T05:23:49.100Z Has data issue: false hasContentIssue false

An update on the efficacy of anti-inflammatory agents for patients with schizophrenia: a meta-analysis

Published online by Cambridge University Press:  23 August 2019

N. Çakici*
Affiliation:
Department of Psychiatry and Amsterdam Neuroscience, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands Antes Center for Mental Health Care, Albrandswaardsedijk 74, 3172 AA, Poortugaal, the Netherlands
N. J. M. van Beveren
Affiliation:
Antes Center for Mental Health Care, Albrandswaardsedijk 74, 3172 AA, Poortugaal, the Netherlands Department of Psychiatry, Erasmus Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, the Netherlands Department of Neuroscience, Erasmus Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
G. Judge-Hundal
Affiliation:
Antes Center for Mental Health Care, Albrandswaardsedijk 74, 3172 AA, Poortugaal, the Netherlands Department of Psychiatry and Biomedical Sciences of Cells and Systems, University Medical Center Groningen, Deusinglaan 2, 9713AW Groningen, the Netherlands
M. M. Koola
Affiliation:
Department of Psychiatry and Behavioral Sciences, George Washington University School of Medicine and Health Sciences, 2300I St NW, Washington, DC 20052, USA
I. E. C. Sommer
Affiliation:
Department of Psychiatry and Biomedical Sciences of Cells and Systems, University Medical Center Groningen, Deusinglaan 2, 9713AW Groningen, the Netherlands
*
Author for correspondence: N. Çakici, E-mail: cakici.n@gmail.com
Rights & Permissions [Opens in a new window]

Abstract

Background

Accumulating evidence shows that a propensity towards a pro-inflammatory status in the brain plays an important role in schizophrenia. Anti-inflammatory drugs might compensate this propensity. This study provides an update regarding the efficacy of agents with some anti-inflammatory actions for schizophrenia symptoms tested in randomized controlled trials (RCTs).

Methods

PubMed, Embase, the National Institutes of Health website (http://www.clinicaltrials.gov), and the Cochrane Database of Systematic Reviews were systematically searched for RCTs that investigated clinical outcomes.

Results

Our search yielded 56 studies that provided information on the efficacy of the following components on symptom severity: aspirin, bexarotene, celecoxib, davunetide, dextromethorphan, estrogens, fatty acids, melatonin, minocycline, N-acetylcysteine (NAC), pioglitazone, piracetam, pregnenolone, statins, varenicline, and withania somnifera extract. The results of aspirin [mean weighted effect size (ES): 0.30; n = 270; 95% CI (CI) 0.06–0.54], estrogens (ES: 0.78; n = 723; CI 0.36–1.19), minocycline (ES: 0.40; n = 946; CI 0.11–0.68), and NAC (ES: 1.00; n = 442; CI 0.60–1.41) were significant in meta-analysis of at least two studies. Subgroup analysis yielded larger positive effects for first-episode psychosis (FEP) or early-phase schizophrenia studies. Bexarotene, celecoxib, davunetide, dextromethorphan, fatty acids, pregnenolone, statins, and varenicline showed no significant effect.

Conclusions

Some, but not all agents with anti-inflammatory properties showed efficacy. Effective agents were aspirin, estrogens, minocycline, and NAC. We observed greater beneficial results on symptom severity in FEP or early-phase schizophrenia.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © The Author(s) 2019

Introduction

The pathophysiology of schizophrenia is still not completely understood, but there is accumulating evidence that dysregulations in components of the immune system are fundamentally linked to the disease. While genetic associations show that people with schizophrenia on average have an immune system subtly more prone to activation, as expressed e.g. in major histocompatibility complex molecules (Debnath et al., Reference Debnath, Cannon and Venkatasubramanian2013; Mokhtari and Lachman, Reference Mokhtari and Lachman2016), its enhancers (Takao et al., Reference Takao, Kobayashi, Hagihara, Ohira, Shoji, Hattori, Koshimizu, Umemori, Toyama, Nakamura, Kuroiwa, Maeda, Atsuzawa, Esaki, Yamaguchi, Furuya, Takagi, Walton, Hayashi, Suzuki, Higuchi, Usuda, Suhara, Nishi, Matsumoto, Ishii and Miyakawa2013), and complement factor 4 (Sekar et al., Reference Sekar, Bialas, de Rivera, Davis, Hammond, Kamitaki, Tooley, Presumey, Baum, Van Doren, Genovese, Rose, Handsaker, Daly, Carroll, Stevens and McCarroll2016), environmental circumstances that naturally activate the immune system such as prenatal infection, trauma, and stress, may put components of the immune system (i.e. microglia) in an altered state of activity (Brown and Derkits, Reference Brown and Derkits2010; Fineberg and Ellman, Reference Fineberg and Ellman2013). Under such circumstances, microglia and other glia may reduce their neurotrophic function and produce less growth factors such as brain-derived neurotrophic factor (BDNF), leading to decreased proliferation of neurons, resulting in reduced connectivity and, finally, brain tissue degradation. In addition, pruning may be increased by opsonization of synaptic buds with activated complement (Nimgaonkar et al., Reference Nimgaonkar, Prasad, Chowdari, Severance and Yolken2017; Presumey et al., Reference Presumey, Bialas and Carroll2017). Glutamatergic and dopaminergic neurotransmissions are particularly vulnerable for an increased activation of microglia, which can induce or exacerbate positive, negative, and cognitive symptoms of schizophrenia (Muller and Schwarz, Reference Muller and Schwarz2006; Muller and Dursun, Reference Muller and Dursun2011).

Over the years, many studies have presented evidence to support this theory. A schizophrenia genome-wide association study found associations between schizophrenia and certain genes that are involved in immune processes (Schizophrenia Working Group of the Psychiatric Genomics, 2014). Peripheral blood markers, such as BDNF, interleukin (IL)-10, and C-reactive protein (CRP), are associated with cognitive decline in schizophrenia (Liu et al., Reference Liu, Zhang, Xie, Ruan, Xu, Zeng, Messina, Zhao and Fan2018; Man et al., Reference Man, Lv, Du, Yin, Zhu, Zhang, Soares, Yang, Chen and Zhang2018; Misiak et al., Reference Misiak, Stanczykiewicz, Kotowicz, Rybakowski, Samochowiec and Frydecka2018). Interestingly, a recent study identified macrophages on the brain side of the endothelial wall in a subgroup of patients with schizophrenia but not in controls, demonstrating an influx of peripheral immune cells (Cai et al., Reference Cai, Catts, Webster, Galletly, Liu, O'Donnell, Weickert and Weickert2018).

The immune hypothesis readily suggests a possible treatment for those patients with schizophrenia in which the underlying pathophysiology is related to a subtle increase in the activation of microglia. Many medications can decrease the production of pro-inflammatory factors; however, it is not certain whether these agents can induce microglia, astrocytes, and other cells to resume their normal neurotrophic functions (Chew et al., Reference Chew, Fusar-Poli and Schmitz2013; Sommer et al., Reference Sommer, van Westrhenen, Begemann, de Witte, Leucht and Kahn2014). For one frequently used anti-inflammatory drug, minocycline, Sellgren et al. showed that this drug was indeed able to reduce microglia engulfment of complement opsonized synapses in a stem cell model derived from patients (Sellgren et al., Reference Sellgren, Gracias, Watmuff, Biag, Thanos, Whittredge, Fu, Worringer, Brown, Wang, Kaykas, Karmacharya, Goold, Sheridan and Perlis2019). This finding suggests that at least minocycline, but perhaps also other anti-inflammatory drugs, can correct one of the basic mechanisms underlying schizophrenia. Yet, components that work in vitro do not always work in vivo.

In a previous meta-analysis on augmentation with anti-inflammatory medications, we showed beneficial results of aspirin, estrogens, and N-acetylcysteine (NAC) on symptom improvement in patients with schizophrenia (Sommer et al., Reference Sommer, van Westrhenen, Begemann, de Witte, Leucht and Kahn2014), though based on very few studies. However, since the publication of our previous meta-analysis, a substantial number of additional studies have investigated the same and other agents with potential anti-inflammatory properties, which could reinstate the balance between synaptogenesis and pruning in schizophrenia and possibly improve symptoms. We have listed in Table 1 treatments with known anti-inflammatory actions, how well the blood-brain-barrier (BBB) can be crossed, and their actions in the brain. This summary is incomplete, as many nutritional and herbal components also possess anti-inflammatory aspects. Additionally, many psychotropic agents such as antipsychotics, selective serotonin reuptake inhibitors, lithium, and valproate acid also have some anti-inflammatory actions. As shown in Table 1, most anti-inflammatory components have many functions, and their anti-inflammatory actions are just one of them and often not the most important one. Some of these agents have been given to patients with schizophrenia in an attempt to normalize the brain's immune system and to eventually reduce symptoms. Here, we quantitatively summarize all available evidence of drugs with some anti-inflammatory aspects studied in patients with schizophrenia in a double-blind randomized design.

Table 1. Main types of medication with anti-inflammatory actions

BBB, blood-brain barrier; BDNF, brain-derived neurotrophic factor; CNS, central nervous system; COX, cyclooxygenase; CRP, C-reactive protein; IFN, interferon; IL, interleukin; MPO, myeloperoxidase; MTX, methotrexate; NF-κβ, nuclear factor-κβ; NO, nitric oxide; PG, prostaglandin; TNF, tumor necrosis factor.

++, excellent BBB crossing; +/−, lower CNS concentrations than in peripheral blood.

a https://www.drugbank.ca/drugs was also accessed to determine the degree of BBB crossing (access date 5 February 2019)

Methods

Literature search

The literature was systematically reviewed according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses (Moher et al., Reference Moher, Liberati, Tetzlaff and Altman2009). Two independent investigators (N.Ç. and G.E.) systematically searched PubMed, Embase, the National Institutes of Health website (http://www.clinicaltrials.gov), and the Cochrane Database of Systematic Reviews from inception to 9 August 2018. No language or year restrictions were applied. The search strategy used for each database can be found in online Supplementary Material S1.

Inclusion criteria

Consensus on the studies included was reached on the basis of the following criteria:

  1. (1) Randomized, double-blind, placebo-controlled trials regarding augmentation of antipsychotic medication with anti-inflammatory agents.

  2. (2) Patients included had a diagnosis of a schizophrenia spectrum disorder (schizophrenia, schizophreniform disorder, or schizoaffective disorder) according to the diagnostic criteria of the Diagnostic and Statistical Manual of Mental Disorders (DSM-III, DSM-III-R, DSM-IV, and DSM-IV-TR or International Classification of Diseases, 9th or 10th revision). Schizotypal and schizoid personality disorder were not included.

  3. (3) Studies reported information to calculate common effect size (ES) statistics of change scores, i.e. means and standard deviations; exact p, t, or z values; or corresponding authors could supply these data upon request. Studies providing only post-treatment data were not included.

We also included crossover studies to obtain as much information as possible. We excluded antipsychotic, antidepressant, and mood-stabilizing agents because their well-known efficacy on symptom severity would confound the results. Studies that were only published as abstracts were included after contacting the authors for more detailed information. If multiple publications from the same cohort were available, we extracted data from the largest or most recent data set.

Outcome measures

The primary outcome measure was the mean change in total score on the Positive and Negative Syndrome Scale (PANSS) or the Brief Psychiatric Rating Scale (BPRS). We also investigated effects on PANSS positive, PANSS negative, and cognitive test batteries. Data of the last observation carried forward analysis were used when provided. If only data of completer analyses were given, these data were used instead. The quality of the studies was assessed using the Cochrane risk of bias tool for randomized trials (Higgins et al., Reference Higgins, Altman, Gotzsche, Juni, Moher, Oxman, Savovic, Schulz, Weeks and Sterne2011). Two reviewers (N.Ç. and G.E.) independently extracted data from the papers. Disagreements were resolved by discussion or by a third reviewer (I.E.S.).

Statistical analyses

We calculated standardized differences from the mean differences (placebo v. augmentation) of the change score (end of treatment minus baseline) means and standard deviations (Rosenthal, Reference Rosenthal1991). When only exact F or p values for the main effects of the treatment group were presented, these data were used. We calculated standardized mean differences, represented as Hedges' g (Shaddish and Haddock, Reference Shaddish, Haddock, Coopers and Hedges1994), using a random-effects model. Inconsistency across studies was assessed with the I 2 statistic (Higgins et al., Reference Higgins, Thompson, Deeks and Altman2003), with values ⩾50% indicating high heterogeneity, and values between 30% and 50% indicating moderate heterogeneity. Potential publication bias was assessed using the Egger test of the intercept if 10 or more studies were analysed for the same anti-inflammatory therapy and represented diagrammatically with funnel plots (Egger et al., Reference Egger, Davey Smith, Schneider and Minder1997), as recommended by the Cochrane Collaboration (online Supplementary Figs S1–S3) (Higgins and Green, Reference Higgins and Green2008). Subgroup analyses were performed to investigate the effects of anti-inflammatory medication in distinct patient groups, including first-episode psychosis (FEP), early-phase schizophrenia (duration of illness ⩽ 5 years) and chronic schizophrenia (duration of illness > 5 years). Meta-regression of categorical moderators was performed if at least four studies were available. In turn, meta-regression of continuous moderators was performed if at least six studies were available (Fu et al., Reference Fu, Gartlehner, Grant, Shamliyan, Sedrakyan, Wilt, Griffith, Oremus, Raina, Ismaila, Santaguida, Lau and Trikalinos2011). Following this rule, we assessed the effects of the following moderators: study quality, illness duration, treatment duration, treatment dose, and baseline severity score (as measured with the PANSS total). Results of meta-analysis and meta-regression with a p value < 0.05 were considered significant. Results of multiple testing, using the Bonferroni correction (Haynes, Reference Haynes, Dubitzky, Wolkenhauer, Cho and Yokota2013), are presented in addition to uncorrected findings for interpretation of the reader. All analyses were performed using Comprehensive Meta-Analysis version 2.0.

Results

A total of 56 studies were retrieved by our search that fulfilled all inclusion criteria (Fig. 1). These studies provided information on the efficacy of the following agents on the improvement of symptom severity in patients with schizophrenia: aspirin, bexarotene, celecoxib, davunetide, dextromethorphan, estrogens, fatty acids including eicosapentaenoic acids (EPA) and docosahexaenoic acids (DHA), melatonin, minocycline, NAC, pioglitazone, piracetam, pregnenolone, statins, varenicline, and withania somnifera extract (WSE). Figure 2 shows the effect sizes of the effects of anti-inflammatory medication on symptom severity. Effect size estimates for individual studies are provided in online Supplementary Figs S4–S19.

Fig. 1. PRISMA flow diagram of the performed literature search.

Fig. 2. Forest Plot Showing Effect Sizes for Anti-Inflammatory Therapies in Schizophrenia.

Additional study characteristics such as treatment duration and treatment dose are provided in online Supplementary Table S1. A detailed description of the effects of anti-inflammatory agents on positive and negative symptoms can be found in online Supplementary Material S2 and online Supplementary Figs S20–S46. Quality of the studies varied from fair to good quality (online Supplementary Table S2).

Aspirin

Aspirin is an NSAID that modifies cyclooxygenase-2 (COX-2) activity and irreversibly inhibits cyclooxygenase-1 (COX-1), thereby suppressing the production of prostaglandins and thromboxanes, which are involved in the inflammatory process (Roth and Majerus, Reference Roth and Majerus1975; Vane et al., Reference Vane, Bakhle and Botting1998). Aspirin also reduces hypothalamic-pituitary-adrenal axis response (Nye et al., Reference Nye, Hockings, Grice, Torpy, Walters, Crosbie, Wagenaar, Cooper and Jackson1997). The BBB is not readily crossed by aspirin, and aspirin levels in the central nervous system are lower than in peripheral blood (Vasovic et al., Reference Vasovic, Banic, Jakovljevic, Tomic and Milic-Djordjevic2008). Two studies provided 1000 mg aspirin daily to schizophrenia patients in addition to their regular treatment for 3 (Laan et al., Reference Laan, Grobbee, Selten, Heijnen, Kahn and Burger2010) or 4 months (Weiser et al., Reference Weiser, Burshtein and Fodoreanu2012). A significant positive influence on total symptom severity was observed [mean weighted effect size (ES): 0.30; 95% confidence interval (CI) 0.06–0.54; p = 0.014; heterogeneity (I 2) = 0%].

Bexarotene

Bexarotene is an antitumor agent that acts via the nuclear retinoid X receptor (RXR) (Lerner et al., Reference Lerner, Miodownik, Gibel, Sirota, Bush, Elliot, Benatov and Ritsner2013). Activation of RXR has the potential to increase apolipoprotein E, which inhibits the complement pathway (Tousi, Reference Tousi2015; Yin et al., Reference Yin, Ackermann, Ma, Mohanta, Zhang, Li, Nietzsche, Westermann, Peng, Hu, Bontha, Srikakulapu, Beer, Megens, Steffens, Hildner, Halder, Eckstein, Pelisek, Herms, Roeber, Arzberger, Borodovsky, Habenicht, Binder, Weber, Zipfel, Skerka and Habenicht2019). Bexarotene can easily cross the BBB (Tousi, Reference Tousi2015). One study investigated the effects of bexarotene 75 mg/day for 6 weeks on symptom severity in schizophrenia patients. However, bexarotene did not significantly improve symptom severity (ES: 0.37; CI −0.05 to 0.78; I 2 = 0%).

Celecoxib

Celecoxib is also an NSAID and has analgesic and inflammatory actions as well. Celecoxib reduces pain and inflammation by blocking COX-2-mediated vascular permeability, thereby reducing extravasation of pro-inflammatory cells, proteins, and enzymes, which enhance the local inflammatory response and lead to edema (Simon, Reference Simon1999). Celecoxib is a small molecule that can easily cross the BBB (Davies et al., Reference Davies, McLachlan, Day and Williams2000). In all five included studies, a dose of 400 mg was provided to schizophrenia patients, and duration of treatment varied from 5 to 11 weeks (Muller et al., Reference Muller, Riedel, Scheppach, Brandstatter, Sokullu, Krampe, Ulmschneider, Engel, Moller and Schwarz2002, Reference Muller, Krause, Dehning, Musil, Schennach-Wolff, Obermeier, Moller, Klauss, Schwarz and Riedel2010; Rappard and Muller, Reference Rappard and Muller2004; Rapaport et al., Reference Rapaport, Delrahim, Bresee, Maddux, Ahmadpour and Dolnak2005; Akhondzadeh et al., Reference Akhondzadeh, Tabatabaee, Amini, Ahmadi Abhari, Abbasi and Behnam2007). We observed heterogeneous results, ranging from strong positive to strong negative effects of celecoxib as augmentation therapy. The effects of celecoxib on the symptom severity was not significant (ES: 0.15; CI −0.67 to 0.96), and heterogeneity was high (I 2 = 93%).

Davunetide

Davunetide is the smallest active element from the activity-dependent neuroprotective protein, which can readily enter the BBB from the blood (Quintana et al., Reference Quintana, Zaltzman, Fernandez-Montesinos, Herrera, Gozes, Cohen and Pozo2006). Davunetide can downregulate key inflammatory cytokines (Quintana et al., Reference Quintana, Zaltzman, Fernandez-Montesinos, Herrera, Gozes, Cohen and Pozo2006). We included one study that provided davunetide (5 or 30 mg daily) as augmentation therapy to patients with chronic schizophrenia for 3 months (Javitt et al., Reference Javitt, Buchanan, Keefe, Kern, McMahon, Green, Lieberman, Goff, Csernansky, McEvoy, Jarskog, Seidman, Gold, Kimhy, Nolan, Barch, Ball, Robinson and Marder2012). Neither dose improved symptom severity (ES: −0.24; CI −0.65 to 0.19; I 2 = 0%).

Dextromethorphan

Dextromethorphan, an antitussive drug, has neuroprotective and anti-inflammatory effects by inhibiting overactivation of microglia (Zhang et al., Reference Zhang, Wang, Qin, Gao, Wilson, Ali, Zhang, Hong and Liu2004). One study provided 60 mg dextromethorphan daily in addition to standard treatment to patients with schizophrenia for 11 weeks (Lee et al., Reference Lee, Chen, Chang, Chen, Huang, Tzeng, Wang, Lee, Wang, Chen, Yang, Hong and Lu2015). Dextromethorphan did not improve symptom severity (ES: 0.11; CI −0.29 to 0.52; I 2 = 0%).

EPA and DHA fatty acids

Fatty acids, especially EPA and DHA fatty acids, have several mild anti-inflammatory effects, such as decreasing levels of serum IL-1β, tumor necrosis factor alpha (TNF-α) and interferon-γ levels, and neuroprotective effects (Solfrizzi et al., Reference Solfrizzi, Frisardi, Capurso, D'Introno, Colacicco, Vendemiale, Capurso and Panza2010; Calder, Reference Calder2012). Fatty acids also enhance synaptic plasticity and membrane fluidity and affect dopaminergic, serotonergic, and glutamatergic neurotransmission (Glantz and Lewis, Reference Glantz and Lewis2000; Calder, Reference Calder2012). Eleven studies were included, of which seven studies added EPA, one study added DHA, and four studies added omega-3 fatty acids (i.e. combination of EPA and DHA) to antipsychotic treatment for patients with schizophrenia (Fenton et al., Reference Fenton, Dickerson, Boronow, Hibbeln and Knable2001; Peet et al., Reference Peet, Brind, Ramchand, Shah and Vankar2001; Peet and Horrobin, Reference Peet and Horrobin2002; Emsley et al., Reference Emsley, Myburgh, Oosthuizen and van Rensburg2002, Reference Emsley, Niehaus, Koen, Oosthuizen, Turner, Carey, van Rensburg, Maritz and Murck2006, Reference Emsley, Chiliza, Asmal, du Plessis, Phahladira, van Niekerk, van Rensburg and Harvey2014; Berger et al., Reference Berger, Proffitt, McConchie, Yuen, Wood, Amminger, Brewer and McGorry2007; Bentsen et al., Reference Bentsen, Osnes, Refsum, Solberg and Bohmer2013; Jamilian et al., Reference Jamilian, Solhi and Jamilian2014; Boskovic et al., Reference Boskovic, Vovk, Koprivsek, Plesnicar and Grabnar2016; Pawelczyk et al., Reference Pawelczyk, Grancow-Grabka, Kotlicka-Antczak, Trafalska and Pawelczyk2016). Daily treatment doses of fatty acids varied (EPA 0.5 g to 4 g, DHA 2 g, omega-3 0.4 g to 2.2 g) as did treatment duration across the studies (8 weeks to 2 years). We observed a trend toward beneficial results for treatment with EPA and/or DHA fatty acids (ES: 0.19; CI −0.02 to 0.40; p = 0.075; I 2 = 41%), without indication of publication bias (Egger test p = 0.45). One study reported a large negative ES of −0.64 and was regarded as an outlier in an additional analysis (Bentsen et al., Reference Bentsen, Osnes, Refsum, Solberg and Bohmer2013). Exclusion of this outlier yielded a mean weighted ES of 0.23, which was significant (CI 0.05–0.41; p = 0.012; I 2 = 9%). Subgroup analysis showed a trend toward beneficial effects for FEP patients (ES: 0.31; CI −0.02 to 0.64; p = 0.064) (online Supplementary Table S3).

Estrogens

Estrogens, especially 17β-estradiol, have immunomodulatory effects by, e.g. regulating innate immune signalling pathways and modulating inflammatory elements such as cytokines (Medina-Estrada et al., Reference Medina-Estrada, Alva-Murillo, Lopez-Meza and Ochoa-Zarzosa2018). Other properties of estrogens include reducing antioxidative stress, controlling energy balance and glucose homeostasis, and influencing dopaminergic neurotransmission (Liu et al., Reference Liu, Fan, Zhao, Luo, Li, Li and Le2005). Eleven studies provided estrogen as augmentation therapy for patients with schizophrenia (Kulkarni et al., Reference Kulkarni, Riedel, de Castella, Fitzgerald, Rolfe, Taffe and Burger2001, Reference Kulkarni, de Castella, Fitzgerald, Gurvich, Bailey, Bartholomeusz and Burger2008, Reference Kulkarni, de Castella, Headey, Marston, Sinclair, Lee, Gurvich, Fitzgerald and Burger2011, Reference Kulkarni, Gavrilidis, Gwini, Worsley, Grigg, Warren, Gurvich, Gilbert, Berk and Davis2016; Akhondzadeh et al., Reference Akhondzadeh, Nejatisafa, Amini, Mohammadi, Larijani, Kashani, Raisi and Kamalipour2003; Louza et al., Reference Louza, Marques, Elkis, Bassitt, Diegoli and Gattaz2004; Ghafari et al., Reference Ghafari, Fararouie, Shirazi, Farhangfar, Ghaderi and Mohammadi2013; Kianimehr et al., Reference Kianimehr, Fatehi, Hashempoor, Khodaei-Ardakani, Rezaei, Nazari, Kashani and Akhondzadeh2014; Khodaie-Ardakani et al., Reference Khodaie-Ardakani, Khosravi, Zarinfard, Nejati, Mohsenian, Tabrizi and Akhondzadeh2015; Usall et al., Reference Usall, Huerta-Ramos, Labad, Cobo, Nunez, Creus, Pares, Cuadras, Franco, Miquel, Reyes and Roca2016; Weiser et al., Reference Weiser, Levi, Burshtein, Hagin, Matei, Podea, Miclutia, Tiugan, Pacala, Grecu, Noy, Zamora and Davis2017). Nine studies included only females, and two studies included only males (Kianimehr et al., Reference Kianimehr, Fatehi, Hashempoor, Khodaei-Ardakani, Rezaei, Nazari, Kashani and Akhondzadeh2014; Khodaie-Ardakani et al., Reference Khodaie-Ardakani, Khosravi, Zarinfard, Nejati, Mohsenian, Tabrizi and Akhondzadeh2015). Four studies applied (ethinyl) estradiol (Kulkarni et al., Reference Kulkarni, Riedel, de Castella, Fitzgerald, Rolfe, Taffe and Burger2001, Reference Kulkarni, de Castella, Fitzgerald, Gurvich, Bailey, Bartholomeusz and Burger2008, Reference Kulkarni, de Castella, Headey, Marston, Sinclair, Lee, Gurvich, Fitzgerald and Burger2011; Akhondzadeh et al., Reference Akhondzadeh, Nejatisafa, Amini, Mohammadi, Larijani, Kashani, Raisi and Kamalipour2003), two studies applied conjugated estrogen (Louza et al., Reference Louza, Marques, Elkis, Bassitt, Diegoli and Gattaz2004; Ghafari et al., Reference Ghafari, Fararouie, Shirazi, Farhangfar, Ghaderi and Mohammadi2013), and five studies applied raloxifene, a selective estrogen receptor modulator (Kianimehr et al., Reference Kianimehr, Fatehi, Hashempoor, Khodaei-Ardakani, Rezaei, Nazari, Kashani and Akhondzadeh2014; Khodaie-Ardakani et al., Reference Khodaie-Ardakani, Khosravi, Zarinfard, Nejati, Mohsenian, Tabrizi and Akhondzadeh2015; Kulkarni et al., Reference Kulkarni, Gavrilidis, Gwini, Worsley, Grigg, Warren, Gurvich, Gilbert, Berk and Davis2016; Usall et al., Reference Usall, Huerta-Ramos, Labad, Cobo, Nunez, Creus, Pares, Cuadras, Franco, Miquel, Reyes and Roca2016; Weiser et al., Reference Weiser, Levi, Burshtein, Hagin, Matei, Podea, Miclutia, Tiugan, Pacala, Grecu, Noy, Zamora and Davis2017). Estrogen doses ranged from 0.05 mg per day (patch) to 2 mg per day (orally), and raloxifene doses varied from 60 mg to 120 mg per day (orally). One study reported a large ES of 3.7 and was regarded as an outlier (Ghafari et al., Reference Ghafari, Fararouie, Shirazi, Farhangfar, Ghaderi and Mohammadi2013). Exclusion of this outlier yielded a mean weighted ES of 0.57, which was significant (CI 0.25–0.90; p = 0.001; I 2 = 74%). Indication of publication bias was found (Egger test p = 0.001). A significant ES was also found when we restricted analyses to female studies only (ES: 0.52; CI 0.18–0.87; p = 0.003; I 2 = 72%).

Melatonin

Melatonin is a multifunctional hormone largely derived from the pineal gland at night under normal light and dark conditions. It is an antioxidant and also a widespread anti-inflammatory molecule, modulating both pro- and anti-inflammatory cytokines, which can easily pass the BBB (Favero et al., Reference Favero, Franceschetti, Bonomini, Rodella and Rezzani2017). One study investigated the effects of adding 3 mg melatonin daily to regular antipsychotic treatment for patients with schizophrenia for 8 weeks (Modabbernia et al., Reference Modabbernia, Heidari, Soleimani, Sobhani, Roshan, Taslimi, Ashrafi and Modabbernia2014). Melatonin showed significant beneficial results on decreasing symptom severity in schizophrenia (ES: 2.82; CI 1.91–3.74; p < 0.001; I 2 = 0%).

Minocycline

Minocycline is a broad-spectrum tetracycline antibiotic that has strong inhibitory effects on microglia cells and can easily cross the BBB (Watabe et al., Reference Watabe, Kato, Monji, Horikawa and Kanba2012). Ten studies assessed the effect of minocycline augmentation therapy for schizophrenia patients (Levkovitz et al., Reference Levkovitz, Mendlovich, Riwkes, Braw, Levkovitch-Verbin, Gal, Fennig, Treves and Kron2010; Chaudhry et al., Reference Chaudhry, Hallak, Husain, Minhas, Stirling, Richardson, Dursun, Dunn and Deakin2012; Ghanizadeh et al., Reference Ghanizadeh, Dehbozorgi, OmraniSigaroodi and Rezaei2014; Khodaie-Ardakani et al., Reference Khodaie-Ardakani, Mirshafiee, Farokhnia, Tajdini, Hosseini, Modabbernia, Rezaei, Salehi, Yekehtaz, Ashrafi, Tabrizi and Akhondzadeh2014; Liu et al., Reference Liu, Guo, Wu, Ou, Zheng, Zhang, Xie, Zhang, Yang, Yang, Yang, Ruan, Zeng, Xu and Zhao2014; Chaves et al., Reference Chaves, Marque, Maia-de-Oliveira, Wichert-Ana, Ferrari, Santos, Araujo, Machado-de-Sousa, Bressan, Elkis, Crippa, Guimaraes, Zuardi, Baker, Dursun and Hallak2015; Kelly et al., Reference Kelly, Sullivan, McEvoy, McMahon, Wehring, Gold, Liu, Warfel, Vyas, Richardson, Fischer, Keller, Koola, Feldman, Russ, Keefe, Osing, Hubzin, August, Walker and Buchanan2015; Deakin et al., Reference Deakin, Suckling, Barnes, Byrne, Chaudhry, Dazzan, Drake, Giordano, Husain, Jones, Joyce, Knox, Krynicki, Lawrie, Lewis, Lisiecka-Ford, Nikkheslat, Pariante, Smallman, Watson, Williams, Upthegrove and Dunn2018; Zhang et al., Reference Zhang, Zheng, Wu, Zhu, Kosten, Zhang and Zhao2018; Weiser et al., Reference Weiser, Levi, Burshtein, Chirita, Cirjaliu, Gonen, Yolken, Davidson, Zamora and Davis2019). The daily treatments doses varied from 100 to 300 mg, and the duration of treatment was relatively long, ranging from 2 to 12 months. Minocycline treatment in addition to regular antipsychotic treatment showed significantly beneficial results on symptom severity (ES: 0.40; CI 0.11–0.68; p = 0.007; I 2 = 77%), with an indication of publication bias (Egger test p < 0.001). One study reported a large negative ES of −0.24 (Deakin et al., Reference Deakin, Suckling, Barnes, Byrne, Chaudhry, Dazzan, Drake, Giordano, Husain, Jones, Joyce, Knox, Krynicki, Lawrie, Lewis, Lisiecka-Ford, Nikkheslat, Pariante, Smallman, Watson, Williams, Upthegrove and Dunn2018). Excluding this study from the analysis yielded a mean weighted ES of 0.47 (CI 0.18–0.76; p = 0.002; I 2 = 72%). Subgroup analysis showed a trend toward positive effects for patients with early-phase schizophrenia (ES: 0.38; CI −0.02 to 0.78; p = 0.060) (online Supplementary Table S3).

N-acetylcysteine

NAC has evident anti-inflammatory properties and can modulate immune functions during the inflammatory response by inhibiting TNF-α, IL-1β, and IL-6 (Palacio et al., Reference Palacio, Markert and Martinez2011). NAC can also easily pass the BBB (Farr et al., Reference Farr, Poon, Dogrukol-Ak, Drake, Banks, Eyerman, Butterfield and Morley2003). Five studies investigated the effects of NAC augmentation therapy on symptom severity of patients with schizophrenia (Berk et al., Reference Berk, Copolov, Dean, Lu, Jeavons, Schapkaitz, Anderson-Hunt, Judd, Katz, Katz, Ording-Jespersen, Little, Conus, Cuenod, Do and Bush2008; Farokhnia et al., Reference Farokhnia, Azarkolah, Adinehfar, Khodaie-Ardakani, Hosseini, Yekehtaz, Tabrizi, Rezaei, Salehi, Sadeghi, Moghadam, Gharibi, Mirshafiee and Akhondzadeh2013; Zhang et al., Reference Zhang, Chen and Jian-rui2015; Breier et al., Reference Breier, Liffick, Hummer, Vohs, Yang, Mehdiyoun, Visco, Metzler, Zhang and Francis2018; Sepehrmanesh et al., Reference Sepehrmanesh, Heidary, Akasheh, Akbari and Heidary2018). Only one of those studies restricted inclusion to FEP patients only (Zhang et al., Reference Zhang, Chen and Jian-rui2015). Treatment doses varied from 600 mg to 3600 mg, and duration of treatment varied from 8 to 52 weeks. NAC as augmentation therapy had significant beneficial effects on decreasing symptom severity in patients with schizophrenia compared with controls (ES: 1.00; CI 0.60–1.41; p < 0.001; I 2 = 75%). Subgroup analysis showed that augmentation therapy with NAC is beneficial in all illness stages, including FEP which yielded the largest ES (ES: 1.42; CI 1.02–1.81; p < 0.001), early-phase schizophrenia (ES: 0.98; CI 0.45–1.51; p < 0.001), and chronic schizophrenia (ES: 0.44; CI 0.11–0.77; p = 0.010) (online Supplementary Table S3).

Pioglitazone

Pioglitazone is an antidiabetic agent with antioxidant and anti-inflammatory actions (Iranpour et al., Reference Iranpour, Zandifar, Farokhnia, Goguol, Yekehtaz, Khodaie-Ardakani, Salehi, Esalatmanesh, Zeionoddini, Mohammadinejad, Zeinoddini and Akhondzadeh2016), and it can cross the BBB (Grommes et al., Reference Grommes, Karlo, Caprariello, Blankenship, Dechant and Landreth2013). One study provided 30 mg pioglitazone daily in addition to standard treatment for 8 weeks to patients with schizophrenia (Iranpour et al., Reference Iranpour, Zandifar, Farokhnia, Goguol, Yekehtaz, Khodaie-Ardakani, Salehi, Esalatmanesh, Zeionoddini, Mohammadinejad, Zeinoddini and Akhondzadeh2016). Pioglitazone showed significant beneficial results on reducing symptom severity (ES: 0.79; CI 0.17–1.41; p = 0.012; I 2 = 0%).

Piracetam

Piracetam is a nootropic analgesic agent and has anti-inflammatory effects. It can reduce TNF-α, IL-1β, and myeloperoxidase. There is some evidence that piracetam can cross the BBB (Brust, Reference Brust1989). One study provided 3200 mg piracetam in addition to regular antipsychotic treatment for 8 weeks to schizophrenia patients (Noorbala et al., Reference Noorbala, Akhondzadeh, Davari-Ashtiani and Amini-Nooshabadi1999). A significant positive influence on total symptom severity was observed (ES: 0.77; CI 0.05 to 1.50; p = 0.036; I 2 = 0%).

Pregnenolone

Pregnenolone is a steroid hormone precursor that regulates neuron growth and cerebral BDNF levels (Naert et al., Reference Naert, Maurice, Tapia-Arancibia and Givalois2007; Murugan et al., Reference Murugan, Jakka, Namani, Mujumdar and Radhakrishnan2019). Pregnenolone is also an anti-inflammatory molecule that can maintain immune homeostasis in various inflammatory conditions (Murugan et al., Reference Murugan, Jakka, Namani, Mujumdar and Radhakrishnan2019). Pregnenolone can readily cross the BBB (Sripada et al., Reference Sripada, Marx, King, Rampton, Ho and Liberzon2013). One study was included that added 50 mg pregnenolone to standard treatment for early-phase schizophrenia patients for 8 weeks (Ritsner et al., Reference Ritsner, Bawakny and Kreinin2014). For this study, we observed no beneficial effects on the symptom severity (ES: 0.16; CI −0.34 to 0.67; I 2 = 0%).

Statins

Statins are usually provided as primary or secondary prevention of cardiovascular diseases. Statins also have anti-inflammatory effects by reducing atherogenesis and, concomitantly, inflammation (Pearson et al., Reference Pearson, Ballantyne, Veltri, Shah, Bird, Lin, Rosenberg and Tershakovec2009). Statins can reduce levels of CRP and IL-6, and improve insulin resistance (Ridker et al., Reference Ridker, Rifai, Pfeffer, Sacks and Braunwald1999; Guclu et al., Reference Guclu, Ozmen, Hekimsoy and Kirmaz2004; Asanuma et al., Reference Asanuma, Oeser, Stanley, Bailey, Shintani and Stein2008). The fat-soluble statins can easily cross the BBB (Sierra et al., Reference Sierra, Ramos, Molina, Esteo, Vazquez and Burgos2011). Two studies provided statins in addition to regular antipsychotic treatment for patients with schizophrenia (Vincenzi et al., Reference Vincenzi, Stock, Borba, Cleary, Oppenheim, Petruzzi, Fan, Copeland, Freudenreich, Cather and Henderson2014; Tajik-Esmaeeli et al., Reference Tajik-Esmaeeli, Moazen-Zadeh, Abbasi, Shariat, Rezaei, Salehi and Akhondzadeh2017). Tajik-Esmaeeli et al. applied 40 mg simvastatin daily for 8 weeks and Vincenzi et al. applied 40 mg pravastatin for 12 weeks. However, beneficial effects on symptom severity were not observed (ES: 0.50; CI −0.25 to 1.25; I 2 = 78%).

Varenicline

Varenicline is a high-affinity partial agonist at α7 nicotinic acetylcholine receptors (nAChRs) and is used to treat nicotine addiction. Varenicline can readily cross the BBB (Kurosawa et al., Reference Kurosawa, Higuchi, Okura, Kobayashi, Kusuhara and Deguchi2017). Activation of the vagus nerve reduces the production of pro-inflammatory cytokines from macrophages, such as TNF-α, in the spleen through a mechanism dependent on nAChRs (Rosas-Ballina and Tracey, Reference Rosas-Ballina and Tracey2009). It has been shown that varenicline administration reduces brain inflammation and promotes recovery of function following experimental stroke (Chen et al., Reference Chen, Bennet and McGregor2017). Two studies provided varenicline in addition to antipsychotic treatment for patients with schizophrenia (Hong et al., Reference Hong, Thaker, McMahon, Summerfelt, Rachbeisel, Fuller, Wonodi, Buchanan, Myers, Heishman, Yang and Nye2011; Smith et al., Reference Smith, Amiaz, Si, Maayan, Jin, Boules, Sershen, Li, Ren, Liu, Youseff, Lajtha, Guidotti, Weiser and Davis2016). Smith et al. applied 4 mg varenicline daily for 8 weeks and Hong et al. applied 1 mg varenicline for 8 weeks. No beneficial effects were observed on symptom severity (ES: 0.24; CI −0.13 to 0.61; I 2 = 24%).

Withania somnifera extract

WSE, mostly used as a medicinal herb in Ayurvedic medicine, has anti-inflammatory actions (i.e. inhibition of NF-κβ inflammatory signalling pathways and COX-2) (Khan et al., Reference Khan, Ahmad, Bani, Kaul, Suri, Satti, Athar and Qazi2006; Mulabagal et al., Reference Mulabagal, Subbaraju, Rao, Sivaramakrishna, Dewitt, Holmes, Sung, Aggarwal, Tsay and Nair2009). WSE consists of various phytochemicals, of which the effects of 1000 mg withaferin A on symptom severity was investigated in one study for 12 weeks (Chengappa et al., Reference Chengappa, Brar, Gannon and Schlicht2018). WSE with drug ligand withaferin A can readily cross the BBB (Kumar and Patnaik, Reference Kumar and Patnaik2016). A significant positive influence on total symptom severity was observed (ES: 0.81; CI 0.32–1.30; p = 0.001; I 2 = 0%).

Effects of moderators

Meta-regression analysis showed that illness duration, treatment duration, treatment dose, and baseline severity were insignificant predictors of the ES estimates for the effects of augmentation with EPA and/or DHA fatty acids, estrogen and minocycline (online Supplementary Table S3). Study quality was not a significant moderator for the celecoxib, EPA and/or DHA fatty acids, estrogen, minocycline and NAC studies.

Cognition

Eighteen studies investigated the effects of anti-inflammatory agents on cognition (online Supplementary Table S4). Heterogeneity of the cognitive tests used across the studies was too great to make a quantitative review of these effects. Notwithstanding, it seemed that minocycline improved attention, executive functions and memory (Levkovitz et al., Reference Levkovitz, Mendlovich, Riwkes, Braw, Levkovitch-Verbin, Gal, Fennig, Treves and Kron2010; Liu et al., Reference Liu, Guo, Wu, Ou, Zheng, Zhang, Xie, Zhang, Yang, Yang, Yang, Ruan, Zeng, Xu and Zhao2014), whereas davunetide (the 5 mg group) improved verbal learning and memory (Javitt et al., Reference Javitt, Buchanan, Keefe, Kern, McMahon, Green, Lieberman, Goff, Csernansky, McEvoy, Jarskog, Seidman, Gold, Kimhy, Nolan, Barch, Ball, Robinson and Marder2012). NAC (Sepehrmanesh et al., Reference Sepehrmanesh, Heidary, Akasheh, Akbari and Heidary2018) improved attention, memory, and executive functions. However, other studies did not observe any beneficial effects on cognition for minocycline (Chaudhry et al., Reference Chaudhry, Hallak, Husain, Minhas, Stirling, Richardson, Dursun, Dunn and Deakin2012; Kelly et al., Reference Kelly, Sullivan, McEvoy, McMahon, Wehring, Gold, Liu, Warfel, Vyas, Richardson, Fischer, Keller, Koola, Feldman, Russ, Keefe, Osing, Hubzin, August, Walker and Buchanan2015; Deakin et al., Reference Deakin, Suckling, Barnes, Byrne, Chaudhry, Dazzan, Drake, Giordano, Husain, Jones, Joyce, Knox, Krynicki, Lawrie, Lewis, Lisiecka-Ford, Nikkheslat, Pariante, Smallman, Watson, Williams, Upthegrove and Dunn2018; Weiser et al., Reference Weiser, Levi, Burshtein, Chirita, Cirjaliu, Gonen, Yolken, Davidson, Zamora and Davis2019) and NAC (Breier et al., Reference Breier, Liffick, Hummer, Vohs, Yang, Mehdiyoun, Visco, Metzler, Zhang and Francis2018). For statins, only one study investigated the effects of pravastatin on cognition and did not observe any significant effects (Vincenzi et al., Reference Vincenzi, Stock, Borba, Cleary, Oppenheim, Petruzzi, Fan, Copeland, Freudenreich, Cather and Henderson2014). For varenicline, no cognitive improvement was observed by Smith et al. (Reference Smith, Amiaz, Si, Maayan, Jin, Boules, Sershen, Li, Ren, Liu, Youseff, Lajtha, Guidotti, Weiser and Davis2016). For the anti-inflammatory components bexarotene, celecoxib, dextromethorphan, melatonin, pioglitazone, piracetam, pregnenolone, and WSE no data on cognitive effects were reported.

Discussion

In this meta-analysis, we quantitively reviewed the efficacy of various anti-inflammatory medications to reduce symptom severity in patients with schizophrenia. We could include data from 56 studies applying 16 different agents in addition to antipsychotic treatment. The results of aspirin, estrogens, minocycline, and NAC showed significantly better results than placebo in meta-analysis of at least two studies, while pioglitazone, piracetam, and WSE were significant in single studies. Bexarotene, celecoxib, davunetide, dextromethorphan, fatty acids, pregnenolone, statins, and varenicline showed no significant beneficial effects.

Effects on symptom severity of specific components

Aspirin was found to have beneficial effects on symptom severity in our current study. It is important to note that aspirin has broadly active substances, and it is unclear whether the beneficial effects of aspirin are solely due to its anti-inflammatory properties. Celecoxib, which is a more specific anti-inflammatory agent, showed no beneficial effects. Another meta-analysis found that celecoxib improved symptoms in FEP patients but not in chronic patients (Zheng et al., Reference Zheng, Cai, Yang, Ungvari, Ng, Muller, Ning and Xiang2017).

Fatty acids as augmentation therapy for patients with schizophrenia showed borderline significant effects on decreasing symptom severity in the current study. However, the included studies showed great heterogeneity in the methods of treatment. Researchers investigated the addition of different fatty acids (i.e. EPA or DHA) or a combination of fatty acids (i.e. EPA and DHA combined). Furthermore, three research groups added antioxidants to the fatty acids treatment regime (Bentsen et al., Reference Bentsen, Osnes, Refsum, Solberg and Bohmer2013; Emsley et al., Reference Emsley, Chiliza, Asmal, du Plessis, Phahladira, van Niekerk, van Rensburg and Harvey2014; Boskovic et al., Reference Boskovic, Vovk, Koprivsek, Plesnicar and Grabnar2016). So, in fact, several different treatment conditions are investigated under the umbrella term ‘fatty acids augmentation’. We also point out that, considering fatty acids augmentation (without anti-oxidants), the results showed a negative association, but this result was greatly influenced by a substantial outlier. Excluding this outlier showed a positive significant association. Furthermore, we observed that FEP patients might benefit the most from treatment with fatty acids compared with patients with a longer illness duration.

In summary, based on the available data, a clear statement about the efficacy of fatty acids, either alone or in combination with anti-oxidants cannot be made yet. Possibly, fatty acids can be beneficial, but the field is still investigating what specific combination of fatty acids is efficacious, and whether or not antioxidants are beneficial. Further research is warranted before a clear recommendation can be made.

Estrogen augmentation therapy for schizophrenia patients showed beneficial effects for a relatively short duration of treatment (starting at 4 weeks). Estrogens act on different ways in the brain and may cause their beneficial effects by mechanisms that are not related to inflammation (e.g. by affecting angiotensin and neurotransmission) (O'Dell et al., Reference O'Dell, Haire, Palmer, Drymalski, Wees, Blakely, Churchill, Eckhoff, Weaver, Doud, Erikson, Dietz, Olson, Maloley, Klassen and Moore1997; Sanchez et al., Reference Sanchez, Morissette and Di Paolo2012).

Minocycline has strong inhibitory effects on microglia cell activation and may, therefore, be expected to have potential as augmentation therapy for schizophrenia (Inta et al., Reference Inta, Lang, Borgwardt, Meyer-Lindenberg and Gass2017). Microglia activation plays an important role during brain development, but excessive microglia activation is also considered a hallmark of neuroinflammation (Inta et al., Reference Inta, Lang, Borgwardt, Meyer-Lindenberg and Gass2017). Complex variations were found in the complement component 4A (C4A) gene in schizophrenia patients. Human C4 protein is localized to neuronal synapses, axons, dendrites, and cell bodies. These results of high complement activity in the development of schizophrenia could explain the reduced numbers of synapses in the brains of patients with schizophrenia (Sekar et al., Reference Sekar, Bialas, de Rivera, Davis, Hammond, Kamitaki, Tooley, Presumey, Baum, Van Doren, Genovese, Rose, Handsaker, Daly, Carroll, Stevens and McCarroll2016).

In the current meta-analysis, we found a clear positive result on amelioration of symptom severity and especially in early-phase schizophrenia. However, it should be noted that a large negative study was part of our analysis which provided almost 22% of the total amount of patients (Deakin et al., Reference Deakin, Suckling, Barnes, Byrne, Chaudhry, Dazzan, Drake, Giordano, Husain, Jones, Joyce, Knox, Krynicki, Lawrie, Lewis, Lisiecka-Ford, Nikkheslat, Pariante, Smallman, Watson, Williams, Upthegrove and Dunn2018). Deakin and colleagues investigated first-episode patients with an illness duration shorter than 5 years. Minocycline seems to have great beneficial effects on improving negative symptoms in schizophrenia (online Supplementary Fig. S40). We noted that the study population studied by Deakin and colleagues had relatively low baseline levels of PANSS negative symptoms (±17) compared with other studies investigating early-phase schizophrenia patients (>22).

NAC has clear anti-inflammatory and immune-modulating actions. All five studies included in this meta-analysis showed beneficial effects on improving symptom severity. Only one study restricted inclusion to FEP patients and yielded the largest beneficial effects on symptom severity (Zhang et al., Reference Zhang, Chen and Jian-rui2015).

Simvastatin showed beneficial effects on improvement of symptom severity (Tajik-Esmaeeli et al., Reference Tajik-Esmaeeli, Moazen-Zadeh, Abbasi, Shariat, Rezaei, Salehi and Akhondzadeh2017), while pravastatin showed no positive significant effects on symptom severity. The difference can be explained by the fact that simvastatin easily crosses the BBB, while pravastatin does not. We found no significant effects of statins on symptom severity when these two studies were combined. More studies are needed to assess the efficacy of statins, especially of fat-soluble statins on symptom severity in schizophrenia.

Effects on cognition

The variety in cognitive assessment tests across the 18 studies that investigated the effects of anti-inflammatory medication on cognition was large. We observed that minocycline, NAC, and davunetide could have some cognitive enhancing properties but future research is needed.

Side effects

Reconsidering the five agents that showed positive results in a meta-analysis of at least two studies, it is worthwhile to consider the side effects of these anti-inflammatory agents. Aspirin use increases the risk of gastrointestinal bleeding and should, therefore, be combined with gastric protection. This serious side effect does not happen infrequently and, therefore, should be considered and monitored. On the other hand, aspirin also possesses cardioprotective properties, which can be beneficial in schizophrenia patients with metabolic syndrome.

Estrogens are not safe for a longer treatment duration than 1–2 months unless combined with progesterone. Estrogens such as raloxifene are sometimes accompanied by hot flashes and gastrointestinal problems. There are potential risks for the occurrence of thromboembolic events and fatal stroke in women with or at increased risk for cardiovascular disease. Therefore, the clinical risk for thromboembolic events should be evaluated and monitored during treatment (Barrett-Connor et al., Reference Barrett-Connor, Mosca, Collins, Geiger, Grady, Kornitzer, McNabb and Wenger2006; Adomaityte et al., Reference Adomaityte, Farooq and Qayyum2008).

Fatty acids are usually well tolerated. There are some reported side effects during administration such as gastrointestinal effects (e.g. constipation or diarrhea) and infection (e.g. upper respiratory infection). The omega-3 fatty acid and anti-oxidant combination might be beneficial (Bentsen et al., Reference Bentsen, Osnes, Refsum, Solberg and Bohmer2013; Bentsen and Landro, Reference Bentsen and Landro2018).

NAC is a well-tolerated drug that can also be administered during pregnancy. NAC has other beneficial effects in schizophrenia, such as attenuating addiction (Gipson, Reference Gipson2016) and given that it is a free radical scavenger (Markoutsa and Xu, Reference Markoutsa and Xu2017). The NAC-varenicline combination may be beneficial in schizophrenia (Koola, Reference Koola2018).

Minocycline is a tetracyclic antibiotic that can be given to a diverse group of patients with schizophrenia. In the included studies, no serious adverse events were observed in the treatment groups.

Limitations

An important limitation is that many anti-inflammatory augmentation treatment strategies have not been sufficiently investigated. Components with strong anti-inflammatory potency, such as glucocorticosteroids, have not been applied yet to patients with schizophrenia. Also, for most anti-inflammatory medications a limited number of studies was available. Most studies did not stratify schizophrenia patients in subgroups of illness duration. Furthermore, there was an insufficient description of signs of inflammation before the start of anti-inflammatory therapy. For designing future research it would be interesting to investigate whether signs of (low-grade) inflammation before the start of the trials would influence the outcome and degree of inflammation. There is increasing evidence from the biomarker research field that cytokine alterations are already present from disease-onset (Schwarz et al., Reference Schwarz, van Beveren, Ramsey, Leweke, Rothermundt, Bogerts, Steiner, Guest and Bahn2014; Upthegrove et al., Reference Upthegrove, Manzanares-Teson and Barnes2014; van Beveren et al., Reference van Beveren, Schwarz, Noll, Guest, Meijer, de Haan and Bahn2014). It would be interesting for further trials to stratify patients according to the presence of immune alterations and to investigate which inflammatory subtypes would benefit the most from anti-inflammatory therapy. This opens up the way for personalized medicine based on inflammatory markers.

Conclusion

The anti-inflammatory medications aspirin, estrogens, minocycline, and NAC improved symptom severity in patients with schizophrenia. We observed greater beneficial results in early-psychosis studies. Evidence for cognitive improvement is scarce. Taken together, there is evidence for the efficacy of some anti-inflammatory agents on symptom severity in schizophrenia which could confirm the immune hypothesis in schizophrenia, but further studies are still needed.

Supplementary material

The supplementary material for this article can be found at https://doi.org/10.1017/S0033291719001995.

Acknowledgements

We kindly thank I. Grabnar et al., for providing us with their original data. This study was supported by the brain foundation of the Netherlands (care for cognition).

Conflict of interest

The funder had no role in the design and reporting of the study. The authors declare no conflict of interest.

References

Adomaityte, J, Farooq, M and Qayyum, R (2008) Effect of raloxifene therapy on venous thromboembolism in postmenopausal women. A meta-analysis. Thrombosis and Haemostasis 99, 338342.Google Scholar
Akhondzadeh, S, Nejatisafa, AA, Amini, H, Mohammadi, MR, Larijani, B, Kashani, L, Raisi, F and Kamalipour, A (2003) Adjunctive estrogen treatment in women with chronic schizophrenia: a double-blind, randomized, and placebo-controlled trial. Progress in Neuro-Psychopharmacology and Biological Psychiatry 27, 10071012.Google Scholar
Akhondzadeh, S, Tabatabaee, M, Amini, H, Ahmadi Abhari, SA, Abbasi, SH and Behnam, B (2007) Celecoxib as adjunctive therapy in schizophrenia: a double-blind, randomized and placebo-controlled trial. Schizophrenia Research 90, 179185.Google Scholar
Aletaha, D and Smolen, JS (2018) Diagnosis and Management of Rheumatoid Arthritis: A Review. JAMA 320, 13601372.Google Scholar
Asanuma, Y, Oeser, A, Stanley, E, Bailey, DG, Shintani, A and Stein, CM (2008) Effects of C-reactive protein and homocysteine on cytokine production: modulation by pravastatin. Archives of Drug Information 1, 1422.Google Scholar
Azizian, H, Khaksari, M, Asadikaram, G, Sepehri, G and Najafipour, H (2018). Therapeutic effects of tamoxifen on metabolic parameters and cytokines modulation in rat model of postmenopausal diabetic cardiovascular dysfunction: Role of classic estrogen receptors. International Immunopharmacology 65, 190198.Google Scholar
Barrett-Connor, E, Mosca, L, Collins, P, Geiger, MJ, Grady, D, Kornitzer, M, McNabb, MA and Wenger, NK (2006) Effects of raloxifene on cardiovascular events and breast cancer in postmenopausal women. New England Journal of Medicine 355, 125137.Google Scholar
Benros, ME, Mortensen, PB and Eaton, WW (2012) Autoimmune diseases and infections as risk factors for schizophrenia. Annals of the New York Academy of Sciences 1262, 5666.Google Scholar
Bentsen, H and Landro, NI (2018) Neurocognitive effects of an omega-3 fatty acid and vitamins E + C in schizophrenia: a randomised controlled trial. Prostaglandins, Leukotrienes, and Essential Fatty Acids 136, 5766.Google Scholar
Bentsen, H, Osnes, K, Refsum, H, Solberg, DK and Bohmer, T (2013) A randomized placebo-controlled trial of an omega-3 fatty acid and vitamins E + C in schizophrenia. Translational Psychiatry 3, e335.Google Scholar
Berger, GE, Proffitt, TM, McConchie, M, Yuen, H, Wood, SJ, Amminger, GP, Brewer, W and McGorry, PD (2007) Ethyl-eicosapentaenoic acid in first-episode psychosis: a randomized, placebo-controlled trial. Journal of Clinical Psychiatry 68, 18671875.Google Scholar
Berk, M, Copolov, D, Dean, O, Lu, K, Jeavons, S, Schapkaitz, I, Anderson-Hunt, M, Judd, F, Katz, F, Katz, P, Ording-Jespersen, S, Little, J, Conus, P, Cuenod, M, Do, KQ and Bush, AI (2008) N-acetyl cysteine as a glutathione precursor for schizophrenia--a double-blind, randomized, placebo-controlled trial. Biological Psychiatry 64, 361368.Google Scholar
Boskovic, M, Vovk, T, Koprivsek, J, Plesnicar, BK and Grabnar, I (2016) Vitamin E and essential polyunsaturated fatty acids supplementation in schizophrenia patients treated with haloperidol. Nutritional Neuroscience 19, 156161.Google Scholar
Breier, A, Liffick, E, Hummer, TA, Vohs, JL, Yang, Z, Mehdiyoun, NF, Visco, AC, Metzler, E, Zhang, Y and Francis, MM (2018) Effects of 12-month, double-blind N-acetyl cysteine on symptoms, cognition and brain morphology in early phase schizophrenia spectrum disorders. Schizophrenia Research 199, 395402.Google Scholar
Brown, AS and Derkits, EJ (2010) Prenatal infection and schizophrenia: a review of epidemiologic and translational studies. American Journal of Psychiatry 167, 261280.Google Scholar
Brust, P (1989) Reversal of scopolamine-induced alterations of choline transport across the blood-brain barrier by the nootropics piracetam and pramiracetam. Arzneimittel-Forschung 39, 12201222.Google Scholar
Cai, HQ, Catts, VS, Webster, MJ, Galletly, C, Liu, D, O'Donnell, M, Weickert, TW and Weickert, CS (2018) Increased macrophages and changed brain endothelial cell gene expression in the frontal cortex of people with schizophrenia displaying inflammation. Molecular Psychiatry. In Press, Corrected Proof.Google Scholar
Calder, PC (2012) Mechanisms of action of (n-3) fatty acids. Journal of Nutrition 142, 592s599s.Google Scholar
Chan, ES and Cronstein, BN (2010). Methotrexate--how does it really work? Nature Reviews: Rheumatology 6, 175178.Google Scholar
Chaudhry, IB, Hallak, J, Husain, N, Minhas, F, Stirling, J, Richardson, P, Dursun, S, Dunn, G and Deakin, B (2012) Minocycline benefits negative symptoms in early schizophrenia: a randomised double-blind placebo-controlled clinical trial in patients on standard treatment. Journal of Psychopharmacology 26, 11851193.Google Scholar
Chaves, C, Marque, CR, Maia-de-Oliveira, JP, Wichert-Ana, L, Ferrari, TB, Santos, AC, Araujo, D, Machado-de-Sousa, JP, Bressan, RA, Elkis, H, Crippa, JA, Guimaraes, FS, Zuardi, AW, Baker, GB, Dursun, SM and Hallak, JE (2015) Effects of minocycline add-on treatment on brain morphometry and cerebral perfusion in recent-onset schizophrenia. Schizophrenia Research 161, 439445.Google Scholar
Chen, S, Bennet, L and McGregor, AL (2017) Delayed varenicline administration reduces inflammation and improves forelimb use following experimental stroke. Journal of Stroke and Cerebrovascular Diseases 26, 27782787.Google Scholar
Chengappa, KNR, Brar, JS, Gannon, JM and Schlicht, PJ (2018) Adjunctive use of a standardized extract of Withania somnifera (Ashwagandha) to treat symptom exacerbation in schizophrenia: a randomized, double-blind, placebo-controlled study. Journal of Clinical Psychiatry 79, 1.Google Scholar
Chew, LJ, Fusar-Poli, P and Schmitz, T (2013) Oligodendroglial alterations and the role of microglia in white matter injury: relevance to schizophrenia. Developmental Neuroscience 35, 102129.Google Scholar
Davies, NM, McLachlan, AJ, Day, RO and Williams, KM (2000) Clinical pharmacokinetics and pharmacodynamics of celecoxib: a selective cyclo-oxygenase-2 inhibitor. Clinical Pharmacokinetics 38, 225242.Google Scholar
Deakin, B, Suckling, J, Barnes, TRE, Byrne, K, Chaudhry, IB, Dazzan, P, Drake, RJ, Giordano, A, Husain, N, Jones, PB, Joyce, E, Knox, E, Krynicki, C, Lawrie, SM, Lewis, S, Lisiecka-Ford, DM, Nikkheslat, N, Pariante, CM, Smallman, R, Watson, A, Williams, SCR, Upthegrove, R and Dunn, G (2018) The benefit of minocycline on negative symptoms of schizophrenia in patients with recent-onset psychosis (BeneMin): a randomised, double-blind, placebo-controlled trial. The Lancet. Psychiatry 5, 885894.Google Scholar
Debnath, M, Cannon, DM and Venkatasubramanian, G (2013) Variation in the major histocompatibility complex [MHC] gene family in schizophrenia: associations and functional implications. Progress in Neuro-Psychopharmacology and Biological Psychiatry 42, 4962.Google Scholar
Dodd, S, Maes, M, Anderson, G, Dean, OM, Moylan, S and Berk, M (2013) Putative neuroprotective agents in neuropsychiatric disorders. Progress in Neuro-Psychopharmacology and Biological Psychiatry 42, 135145.Google Scholar
Egger, M, Davey Smith, G, Schneider, M and Minder, C (1997) Bias in meta-analysis detected by a simple, graphical test. British Medical Journal 315, 629634.Google Scholar
Emsley, R, Myburgh, C, Oosthuizen, P and van Rensburg, SJ (2002) Randomized, placebo-controlled study of ethyl-eicosapentaenoic acid as supplemental treatment in schizophrenia. American Journal of Psychiatry 159, 15961598.Google Scholar
Emsley, R, Niehaus, DJ, Koen, L, Oosthuizen, PP, Turner, HJ, Carey, P, van Rensburg, SJ, Maritz, JS and Murck, H (2006) The effects of eicosapentaenoic acid in tardive dyskinesia: a randomized, placebo-controlled trial. Schizophrenia Research 84, 112120.Google Scholar
Emsley, R, Chiliza, B, Asmal, L, du Plessis, S, Phahladira, L, van Niekerk, E, van Rensburg, SJ and Harvey, BH (2014) A randomized, controlled trial of omega-3 fatty acids plus an antioxidant for relapse prevention after antipsychotic discontinuation in first-episode schizophrenia. Schizophrenia Research 158, 230235.Google Scholar
Farokhnia, M, Azarkolah, A, Adinehfar, F, Khodaie-Ardakani, MR, Hosseini, SM, Yekehtaz, H, Tabrizi, M, Rezaei, F, Salehi, B, Sadeghi, SM, Moghadam, M, Gharibi, F, Mirshafiee, O and Akhondzadeh, S (2013) N-acetylcysteine as an adjunct to risperidone for treatment of negative symptoms in patients with chronic schizophrenia: a randomized, double- blind, placebo-controlled study. Clinical Neuropharmacology 36, 185192.Google Scholar
Farr, SA, Poon, HF, Dogrukol-Ak, D, Drake, J, Banks, WA, Eyerman, E, Butterfield, DA and Morley, JE (2003) The antioxidants alpha-lipoic acid and N-acetylcysteine reverse memory impairment and brain oxidative stress in aged SAMP8 mice. Journal of Neurochemistry 84, 11731183.Google Scholar
Favero, G, Franceschetti, L, Bonomini, F, Rodella, LF and Rezzani, R (2017) Melatonin as an anti-inflammatory agent modulating inflammasome activation. International Journal of Endocrinology 2017, 1835195.Google Scholar
Fenton, WS, Dickerson, F, Boronow, J, Hibbeln, JR and Knable, M (2001) A placebo-controlled trial of omega-3 fatty acid (ethyl eicosapentaenoic acid) supplementation for residual symptoms and cognitive impairment in schizophrenia. American Journal of Psychiatry 158, 20712074.Google Scholar
Ferreira, AP, Pasin, JS, Saraiva, AL, Ratzlaff, V, Rossato, MF, Andrighetto, R, Rubin, MA, Ferreira, J and Mello, CF (2012) N-acetylcysteine prevents baker's-yeast-induced inflammation and fever. Inflammation Research 61, 103112.Google Scholar
Fineberg, AM and Ellman, LM (2013) Inflammatory cytokines and neurological and neurocognitive alterations in the course of schizophrenia. Biological Psychiatry 73, 951966.Google Scholar
Fu, R, Gartlehner, G, Grant, M, Shamliyan, T, Sedrakyan, A, Wilt, TJ, Griffith, L, Oremus, M, Raina, P, Ismaila, A, Santaguida, P, Lau, J and Trikalinos, TA (2011) Conducting quantitative synthesis when comparing medical interventions: AHRQ and the Effective Health Care Program. Journal of Clinical Epidemiology 64, 11871197.Google Scholar
Ghafari, E, Fararouie, M, Shirazi, HG, Farhangfar, A, Ghaderi, F and Mohammadi, A (2013) Combination of estrogen and antipsychotics in the treatment of women with chronic schizophrenia: a double-blind, randomized, placebo-controlled clinical trial. Clinical Schizophrenia & Related Psychoses 6, 172176.Google Scholar
Ghanizadeh, A, Dehbozorgi, S, OmraniSigaroodi, M and Rezaei, Z (2014) Minocycline as add-on treatment decreases the negative symptoms of schizophrenia; a randomized placebo-controlled clinical trial. Recent Patents on Inflammation & Allergy Drug Discovery 8, 211215.Google Scholar
Gipson, CD (2016) Treating addiction: unraveling the relationship between N-acetylcysteine, glial glutamate transport, and behavior. Biological Psychiatry 80, e11e12.Google Scholar
Glantz, LA and Lewis, DA (2000) Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia. Archives of General Psychiatry 57, 6573.Google Scholar
Grommes, C, Karlo, JC, Caprariello, A, Blankenship, D, Dechant, A and Landreth, GE (2013) The PPARgamma agonist pioglitazone crosses the blood-brain barrier and reduces tumor growth in a human xenograft model. Cancer Chemotherapy and Pharmacology 71, 929936.Google Scholar
Guclu, F, Ozmen, B, Hekimsoy, Z and Kirmaz, C (2004) Effects of a statin group drug, pravastatin, on the insulin resistance in patients with metabolic syndrome. Biomedicine and Pharmacotherapy 58, 614618.Google Scholar
Haynes, W (2013) Bonferroni correction. In Dubitzky, W, Wolkenhauer, O, Cho, KH and Yokota, H (eds), Encyclopedia of Systems Biology. New York: Springer, pp. 154154.Google Scholar
Higgins, JPT and Green, S (2008) Cochrane Collaboration: Cochrane Handbook for Systematic Reviews of Interventions. Chichester, England: Wiley-Blackwell.Google Scholar
Higgins, JP, Thompson, SG, Deeks, JJ and Altman, DG (2003) Measuring inconsistency in meta-analyses. British Medical Journal 327, 557560.Google Scholar
Higgins, JP, Altman, DG, Gotzsche, PC, Juni, P, Moher, D, Oxman, AD, Savovic, J, Schulz, KF, Weeks, L and Sterne, JA (2011) The Cochrane Collaboration's tool for assessing risk of bias in randomised trials. British Medical Journal 343, d5928.Google Scholar
Hong, LE, Thaker, GK, McMahon, RP, Summerfelt, A, Rachbeisel, J, Fuller, RL, Wonodi, I, Buchanan, RW, Myers, C, Heishman, SJ, Yang, J and Nye, A (2011) Effects of moderate-dose treatment with varenicline on neurobiological and cognitive biomarkers in smokers and nonsmokers with schizophrenia or schizoaffective disorder. Archives of General Psychiatry 68, 11951206.Google Scholar
Inta, D, Lang, UE, Borgwardt, S, Meyer-Lindenberg, A and Gass, P (2017) Microglia activation and schizophrenia: lessons from the effects of minocycline on postnatal neurogenesis, neuronal survival and synaptic pruning. Schizophrenia Bulletin 43, 493496.Google Scholar
Iranpour, N, Zandifar, A, Farokhnia, M, Goguol, A, Yekehtaz, H, Khodaie-Ardakani, MR, Salehi, B, Esalatmanesh, S, Zeionoddini, A, Mohammadinejad, P, Zeinoddini, A and Akhondzadeh, S (2016) The effects of pioglitazone adjuvant therapy on negative symptoms of patients with chronic schizophrenia: a double-blind and placebo-controlled trial. Human Psychopharmacology 31, 103112.Google Scholar
Jamilian, H, Solhi, H and Jamilian, M (2014) Randomized, placebo-controlled clinical trial of omega-3 as supplemental treatment in schizophrenia. Global Journal of Health Science 6, 103108.Google Scholar
Javitt, DC, Buchanan, RW, Keefe, RS, Kern, R, McMahon, RP, Green, MF, Lieberman, J, Goff, DC, Csernansky, JG, McEvoy, JP, Jarskog, F, Seidman, LJ, Gold, JM, Kimhy, D, Nolan, KS, Barch, DS, Ball, MP, Robinson, J and Marder, SR (2012) Effect of the neuroprotective peptide davunetide (AL-108) on cognition and functional capacity in schizophrenia. Schizophrenia Research 136, 2531.Google Scholar
Kelly, DL, Sullivan, KM, McEvoy, JP, McMahon, RP, Wehring, HJ, Gold, JM, Liu, F, Warfel, D, Vyas, G, Richardson, CM, Fischer, BA, Keller, WR, Koola, MM, Feldman, SM, Russ, JC, Keefe, RS, Osing, J, Hubzin, L, August, S, Walker, TM and Buchanan, RW (2015) Adjunctive minocycline in clozapine-treated schizophrenia patients with persistent symptoms. Journal of Clinical Psychopharmacology 35, 374381.Google Scholar
Khan, B, Ahmad, SF, Bani, S, Kaul, A, Suri, KA, Satti, NK, Athar, M and Qazi, GN (2006) Augmentation and proliferation of T lymphocytes and Th-1 cytokines by Withania somnifera in stressed mice. International Immunopharmacology 6, 13941403.Google Scholar
Khodaie-Ardakani, MR, Mirshafiee, O, Farokhnia, M, Tajdini, M, Hosseini, SM, Modabbernia, A, Rezaei, F, Salehi, B, Yekehtaz, H, Ashrafi, M, Tabrizi, M and Akhondzadeh, S (2014) Minocycline add-on to risperidone for treatment of negative symptoms in patients with stable schizophrenia: randomized double-blind placebo-controlled study. Psychiatry Research 215, 540546.Google Scholar
Khodaie-Ardakani, MR, Khosravi, M, Zarinfard, R, Nejati, S, Mohsenian, A, Tabrizi, M and Akhondzadeh, S (2015) A placebo-controlled study of raloxifene added to risperidone in men with chronic schizophrenia. Acta Medica Iranica 53, 337345.Google Scholar
Kianimehr, G, Fatehi, F, Hashempoor, S, Khodaei-Ardakani, MR, Rezaei, F, Nazari, A, Kashani, L and Akhondzadeh, S (2014) Raloxifene adjunctive therapy for postmenopausal women suffering from chronic schizophrenia: a randomized double-blind and placebo controlled trial. Daru: Journal of Faculty of Pharmacy, Tehran University of Medical Sciences 22, 55.Google Scholar
Koola, MM (2018) Can Nacetylcysteine, varenicline, or the combination prevent psychosis by enhancing mismatch negativity? Schizophrenia Research. 206, 452453.Google Scholar
Kotsch, K, Ulrich, F, Reutzel-Selke, A, Pascher, A, Faber, W, Warnick, P, Hoffman, S, Francuski, M, Kunert, C, Kuecuek, O, Schumacher, G, Wesslau, C, Lun, A, Kohler, S, Weiss, S, Tullius, SG, Neuhaus, P and Pratschke, J (2008) Methylprednisolone therapy in deceased donors reduces inflammation in the donor liver and improves outcome after liver transplantation: a prospective randomized controlled trial. Annals of Surgery 248, 10421050.Google Scholar
Kulkarni, J, Riedel, A, de Castella, AR, Fitzgerald, PB, Rolfe, TJ, Taffe, J and Burger, H (2001) Estrogen - a potential treatment for schizophrenia. Schizophrenia Research 48, 137144.Google Scholar
Kulkarni, J, de Castella, A, Fitzgerald, PB, Gurvich, CT, Bailey, M, Bartholomeusz, C and Burger, H (2008) Estrogen in severe mental illness: a potential new treatment approach. Archives of General Psychiatry 65, 955960.Google Scholar
Kulkarni, J, de Castella, A, Headey, B, Marston, N, Sinclair, K, Lee, S, Gurvich, C, Fitzgerald, PB and Burger, H (2011) Estrogens and men with schizophrenia: is there a case for adjunctive therapy? Schizophrenia Research 125, 278283.Google Scholar
Kulkarni, J, Gavrilidis, E, Gwini, SM, Worsley, R, Grigg, J, Warren, A, Gurvich, C, Gilbert, H, Berk, M and Davis, SR (2016) Effect of adjunctive raloxifene therapy on severity of refractory schizophrenia in women: a randomized clinical trial. JAMA Psychiatry 73, 947954.Google Scholar
Kumar, G and Patnaik, R (2016) Exploring neuroprotective potential of Withania somnifera phytochemicals by inhibition of GluN2B-containing NMDA receptors: an in silico study. Medical Hypotheses 92, 3543.Google Scholar
Kurosawa, T, Higuchi, K, Okura, T, Kobayashi, K, Kusuhara, H and Deguchi, Y (2017) Involvement of proton-coupled organic cation antiporter in varenicline transport at blood-brain barrier of rats and in human brain capillary endothelial cells. Journal of Pharmaceutical Sciences 106, 25762582.Google Scholar
Laan, W, Grobbee, DE, Selten, JP, Heijnen, CJ, Kahn, RS and Burger, H (2010) Adjuvant aspirin therapy reduces symptoms of schizophrenia spectrum disorders: results from a randomized, double-blind, placebo-controlled trial. Journal of Clinical Psychiatry 71, 520527.Google Scholar
Lee, SY, Chen, SL, Chang, YH, Chen, PS, Huang, SY, Tzeng, NS, Wang, LJ, Lee, IH, Wang, TY, Chen, KC, Yang, YK, Hong, JS and Lu, RB (2015) ALDH2 polymorphism, associated with attenuating negative symptoms in patients with schizophrenia treated with add-on dextromethorphan. Journal of Psychiatric Research 69, 5056.Google Scholar
Lerner, V, Miodownik, C, Gibel, A, Sirota, P, Bush, I, Elliot, H, Benatov, R and Ritsner, MS (2013) The retinoid X receptor agonist bexarotene relieves positive symptoms of schizophrenia: a 6-week, randomized, double-blind, placebo-controlled multicenter trial. Journal of Clinical Psychiatry 74, 12241232.Google Scholar
Levkovitz, Y, Mendlovich, S, Riwkes, S, Braw, Y, Levkovitch-Verbin, H, Gal, G, Fennig, S, Treves, I and Kron, S (2010) A double-blind, randomized study of minocycline for the treatment of negative and cognitive symptoms in early-phase schizophrenia. Journal of Clinical Psychiatry 71, 138149.Google Scholar
Liu, X, Fan, XL, Zhao, Y, Luo, GR, Li, XP, Li, R and Le, WD (2005) Estrogen provides neuroprotection against activated microglia-induced dopaminergic neuronal injury through both estrogen receptor-alpha and estrogen receptor-beta in microglia. Journal of Neuroscience Research 81, 653665.Google Scholar
Liu, D, Ahmet, A, Ward, L, Krishnamoorthy, P, Mandelcorn, ED, Leigh, R, Brown, JP, Cohen, A and Kim, H (2013) A practical guide to the monitoring and management of the complications of systemic corticosteroid therapy. Allergy, Asthma, and Clinical Immunology 9, 30.Google Scholar
Liu, F, Guo, X, Wu, R, Ou, J, Zheng, Y, Zhang, B, Xie, L, Zhang, L, Yang, L, Yang, S, Yang, J, Ruan, Y, Zeng, Y, Xu, X and Zhao, J (2014) Minocycline supplementation for treatment of negative symptoms in early-phase schizophrenia: a double blind, randomized, controlled trial. Schizophrenia Research 153, 169176.Google Scholar
Liu, F, Zhang, B, Xie, L, Ruan, Y, Xu, X, Zeng, Y, Messina, L, Zhao, J and Fan, X (2018) Changes in plasma levels of nitric oxide metabolites and negative symptoms after 16-week minocycline treatment in patients with schizophrenia. Schizophrenia Research 199, 390394.Google Scholar
Louza, MR, Marques, AP, Elkis, H, Bassitt, D, Diegoli, M and Gattaz, WF (2004) Conjugated estrogens as adjuvant therapy in the treatment of acute schizophrenia: a double-blind study. Schizophrenia Research 66, 97100.Google Scholar
Man, L, Lv, X, Du, XD, Yin, G, Zhu, X, Zhang, Y, Soares, JC, Yang, XN, Chen, X and Zhang, XY (2018) Cognitive impairments and low BDNF serum levels in first-episode drug-naive patients with schizophrenia. Psychiatry Research 263, 16.Google Scholar
Markoutsa, E and Xu, P (2017) Redox potential-sensitive N-acetyl cysteine-prodrug nanoparticles inhibit the activation of microglia and improve neuronal survival. Molecular Pharmaceutics 14, 15911600.Google Scholar
Medina-Estrada, I, Alva-Murillo, N, Lopez-Meza, JE and Ochoa-Zarzosa, A (2018) Immunomodulatory effects of 17beta-estradiol on epithelial cells during bacterial infections. Journal of Immunology Research 2018, 6098961.Google Scholar
Miller, BJ and Buckley, PF (2016). The Case for Adjunctive Monoclonal Antibody Immunotherapy in Schizophrenia. Psychiatric Clinics of North America 39, 187198.Google Scholar
Misiak, B, Stanczykiewicz, B, Kotowicz, K, Rybakowski, JK, Samochowiec, J and Frydecka, D (2018) Cytokines and C-reactive protein alterations with respect to cognitive impairment in schizophrenia and bipolar disorder: a systematic review. Schizophrenia Research 192, 1629.Google Scholar
Modabbernia, A, Heidari, P, Soleimani, R, Sobhani, A, Roshan, ZA, Taslimi, S, Ashrafi, M and Modabbernia, MJ (2014) Melatonin for prevention of metabolic side-effects of olanzapine in patients with first-episode schizophrenia: randomized double-blind placebo-controlled study. Journal of Psychiatric Research 53, 133140.Google Scholar
Moher, D, Liberati, A, Tetzlaff, J and Altman, DG (2009) Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Journal of Clinical Epidemiology 62, 10061012.Google Scholar
Mokhtari, R and Lachman, HM (2016) The Major Histocompatibility Complex (MHC) in schizophrenia: a review. Journal of Clinical & Cellular Immunology 7, 479.Google Scholar
Mulabagal, V, Subbaraju, GV, Rao, CV, Sivaramakrishna, C, Dewitt, DL, Holmes, D, Sung, B, Aggarwal, BB, Tsay, HS and Nair, MG (2009) Withanolide sulfoxide from Aswagandha roots inhibits nuclear transcription factor-kappa-B, cyclooxygenase and tumor cell proliferation. Phytotherapy Research 23, 987992.Google Scholar
Mulders-Manders, CM, Baas, MC, Molenaar, FM and Simon, A (2017) Peri- and Postoperative Treatment with the Interleukin-1 Receptor Antagonist Anakinra Is Safe in Patients Undergoing Renal Transplantation: Case Series and Review of the Literature. Frontiers in Pharmacology 8, 342.Google Scholar
Muller, N and Schwarz, M (2006) Schizophrenia as an inflammation-mediated dysbalance of glutamatergic neurotransmission. Neurotoxicity Research 10, 131148.Google Scholar
Muller, N and Dursun, SM (2011) Schizophrenia genes, epigenetics and psychoneuroimmunology therapeutics: all make sense now? Journal of Psychopharmacology 25, 713714.Google Scholar
Muller, N, Riedel, M, Scheppach, C, Brandstatter, B, Sokullu, S, Krampe, K, Ulmschneider, M, Engel, RR, Moller, HJ and Schwarz, MJ (2002) Beneficial antipsychotic effects of celecoxib add-on therapy compared to risperidone alone in schizophrenia. American Journal of Psychiatry 159, 10291034.Google Scholar
Muller, N, Krause, D, Dehning, S, Musil, R, Schennach-Wolff, R, Obermeier, M, Moller, HJ, Klauss, V, Schwarz, MJ and Riedel, M (2010) Celecoxib treatment in an early stage of schizophrenia: results of a randomized, double-blind, placebo-controlled trial of celecoxib augmentation of amisulpride treatment. Schizophrenia Research 121, 118124.Google Scholar
Murugan, S, Jakka, P, Namani, S, Mujumdar, V and Radhakrishnan, G (2019) The neurosteroid, pregnenolone promotes degradation of key proteins in the innate immune signalling to suppress inflammation. Journal of Biological Chemistry 294, 45964607.Google Scholar
Naert, G, Maurice, T, Tapia-Arancibia, L and Givalois, L (2007) Neuroactive steroids modulate HPA axis activity and cerebral brain-derived neurotrophic factor (BDNF) protein levels in adult male rats. Psychoneuroendocrinology 32, 10621078.Google Scholar
Navarro, SA, Serafim, KG, Mizokami, SS, Hohmann, MS, Casagrande, R and Verri, WA Jr. (2013) Analgesic activity of piracetam: effect on cytokine production and oxidative stress. Pharmacology, Biochemistry and Behavior 105, 183192.Google Scholar
Nimgaonkar, VL, Prasad, KM, Chowdari, KV, Severance, EG and Yolken, RH (2017) The complement system: a gateway to gene-environment interactions in schizophrenia pathogenesis. Molecular Psychiatry 22, 15541561.Google Scholar
Noorbala, AA, Akhondzadeh, S, Davari-Ashtiani, R and Amini-Nooshabadi, H (1999) Piracetam in the treatment of schizophrenia: implications for the glutamate hypothesis of schizophrenia. Journal of Clinical Pharmacy and Therapeutics 24, 369374.Google Scholar
Nye, EJ, Hockings, GI, Grice, JE, Torpy, DJ, Walters, MM, Crosbie, GV, Wagenaar, M, Cooper, M and Jackson, RV (1997) Aspirin inhibits vasopressin-induced hypothalamic-pituitary-adrenal activity in normal humans. Journal of Clinical Endocrinology and Metabolism 82, 812817.Google Scholar
O'Dell, JR, Haire, CE, Palmer, W, Drymalski, W, Wees, S, Blakely, K, Churchill, M, Eckhoff, PJ, Weaver, A, Doud, D, Erikson, N, Dietz, F, Olson, R, Maloley, P, Klassen, LW and Moore, GF (1997) Treatment of early rheumatoid arthritis with minocycline or placebo: results of a randomized, double-blind, placebo-controlled trial. Arthritis and Rheumatism 40, 842848.Google Scholar
Palacio, JR, Markert, UR and Martinez, P (2011) Anti-inflammatory properties of N-acetylcysteine on lipopolysaccharide-activated macrophages. Inflammation Research 60, 695704.Google Scholar
Pawelczyk, T, Grancow-Grabka, M, Kotlicka-Antczak, M, Trafalska, E and Pawelczyk, A (2016) A randomized controlled study of the efficacy of six-month supplementation with concentrated fish oil rich in omega-3 polyunsaturated fatty acids in first episode schizophrenia. Journal of Psychiatric Research 73, 3444.Google Scholar
Pearson, TA, Ballantyne, CM, Veltri, E, Shah, A, Bird, S, Lin, J, Rosenberg, E and Tershakovec, AM (2009) Pooled analyses of effects on C-reactive protein and low density lipoprotein cholesterol in placebo-controlled trials of ezetimibe monotherapy or ezetimibe added to baseline statin therapy. American Journal of Cardiology 103, 369374.Google Scholar
Peet, M and Horrobin, DF (2002) A dose-ranging exploratory study of the effects of ethyl-eicosapentaenoate in patients with persistent schizophrenic symptoms. Journal of Psychiatric Research 36, 718.Google Scholar
Peet, M, Brind, J, Ramchand, CN, Shah, S and Vankar, GK (2001) Two double-blind placebo-controlled pilot studies of eicosapentaenoic acid in the treatment of schizophrenia. Schizophrenia Research 49, 243251.Google Scholar
Pettit, LK, Varsanyi, C, Tadros, J and Vassiliou, E (2013) Modulating the inflammatory properties of activated microglia with Docosahexaenoic acid and Aspirin. Lipids in Health and Disease 12, 16.Google Scholar
Presumey, J, Bialas, AR and Carroll, MC (2017) Complement system in neural synapse elimination in development and disease. Advances in Immunology 135, 5379.Google Scholar
Quintana, FJ, Zaltzman, R, Fernandez-Montesinos, R, Herrera, JL, Gozes, I, Cohen, IR and Pozo, D (2006) NAP, a peptide derived from the activity-dependent neuroprotective protein, modulates macrophage function. Annals of the New York Academy of Sciences 1070, 500506.Google Scholar
Rapaport, MH, Delrahim, KK, Bresee, CJ, Maddux, RE, Ahmadpour, O and Dolnak, D (2005) Celecoxib augmentation of continuously ill patients with schizophrenia. Biological Psychiatry 57, 15941596.Google Scholar
Rappard, F and Muller, N (2004) Celecoxib add-on does not have beneficial antipsychotic effects over risperidone alone in schizophrenia [abstract]. Neuropsychopharmacology 29, S183S241.Google Scholar
Ridker, PM, Rifai, N, Pfeffer, MA, Sacks, F and Braunwald, E (1999) Long-term effects of pravastatin on plasma concentration of C-reactive protein. The Cholesterol and Recurrent Events (CARE) Investigators. Circulation 100, 230235.Google Scholar
Ritsner, MS, Bawakny, H and Kreinin, A (2014) Pregnenolone treatment reduces severity of negative symptoms in recent-onset schizophrenia: an 8-week, double-blind, randomized add-on two-center trial. Psychiatry and Clinical Neurosciences 68, 432440.Google Scholar
Rosas-Ballina, M and Tracey, KJ (2009) Cholinergic control of inflammation. Journal of Internal Medicine 265, 663679.Google Scholar
Rosenthal, R (1991) Meta-Analytic Procedures for Social Research. Newbury Park, CA: Sage Publications.Google Scholar
Roth, GJ and Majerus, PW (1975) The mechanism of the effect of aspirin on human platelets. I. Acetylation of a particulate fraction protein. Journal of Clinical Investigation 56, 624632.Google Scholar
Sadli, N, Ackland, ML, De Mel, D, Sinclair, AJ and Suphioglu, C (2012). Effects of zinc and DHA on the epigenetic regulation of human neuronal cells. Cellular Physiology and Biochemistry 29, 8798.Google Scholar
Sanchez, MG, Morissette, M and Di Paolo, T (2012) Effect of a chronic treatment with 17beta-estradiol on striatal dopamine neurotransmission and the Akt/GSK3 signaling pathway in the brain of ovariectomized monkeys. Psychoneuroendocrinology 37, 280291.Google Scholar
Schizophrenia Working Group of the Psychiatric Genomics (2014) Biological insights from 108 schizophrenia-associated genetic loci. Nature 511, 421.Google Scholar
Schwarz, E, van Beveren, NJ, Ramsey, J, Leweke, FM, Rothermundt, M, Bogerts, B, Steiner, J, Guest, PC and Bahn, S (2014) Identification of subgroups of schizophrenia patients with changes in either immune or growth factor and hormonal pathways. Schizophrenia Bulletin 40, 787795.Google Scholar
Sekar, A, Bialas, AR, de Rivera, H, Davis, A, Hammond, TR, Kamitaki, N, Tooley, K, Presumey, J, Baum, M, Van Doren, V, Genovese, G, Rose, SA, Handsaker, RE, Daly, MJ, Carroll, MC, Stevens, B and McCarroll, SA (2016) Schizophrenia risk from complex variation of complement component 4. Nature 530, 177183.Google Scholar
Sellgren, CM, Gracias, J, Watmuff, B, Biag, JD, Thanos, JM, Whittredge, PB, Fu, T, Worringer, K, Brown, HE, Wang, J, Kaykas, A, Karmacharya, R, Goold, CP, Sheridan, SD and Perlis, RH (2019) Increased synapse elimination by microglia in schizophrenia patient-derived models of synaptic pruning. Nature Neuroscience 22, 374385.Google Scholar
Sepehrmanesh, Z, Heidary, M, Akasheh, N, Akbari, H and Heidary, M (2018) Therapeutic effect of adjunctive N-acetyl cysteine (NAC) on symptoms of chronic schizophrenia: a double-blind, randomized clinical trial. Progress in Neuro-Psychopharmacology and Biological Psychiatry 82, 289296.Google Scholar
Shaddish, W and Haddock, C (1994) Combining estimates of effect size. In The Handbook of Research Synthesis (ed. Coopers, H. and Hedges, V.), pp. 261281. Sage Publications: New York.Google Scholar
Sierra, S, Ramos, MC, Molina, P, Esteo, C, Vazquez, JA and Burgos, JS (2011) Statins as neuroprotectants: a comparative in vitro study of lipophilicity, blood-brain-barrier penetration, lowering of brain cholesterol, and decrease of neuron cell death. Journal of Alzheimer's Disease 23, 307318.Google Scholar
Simon, LS (1999) Role and regulation of cyclooxygenase-2 during inflammation. American Journal of Medicine 106, 37s42s.Google Scholar
Smith, RC, Amiaz, R, Si, TM, Maayan, L, Jin, H, Boules, S, Sershen, H, Li, C, Ren, J, Liu, Y, Youseff, M, Lajtha, A, Guidotti, A, Weiser, M and Davis, JM (2016) Varenicline effects on smoking, cognition, and psychiatric symptoms in schizophrenia: a double-blind randomized trial. PloS One 11, e0143490.Google Scholar
Solfrizzi, V, Frisardi, V, Capurso, C, D'Introno, A, Colacicco, AM, Vendemiale, G, Capurso, A and Panza, F (2010) Dietary fatty acids in dementia and predementia syndromes: epidemiological evidence and possible underlying mechanisms. Ageing Research Reviews 9, 184199.Google Scholar
Sommer, IE, van Westrhenen, R, Begemann, MJ, de Witte, LD, Leucht, S and Kahn, RS (2014) Efficacy of anti-inflammatory agents to improve symptoms in patients with schizophrenia: an update. Schizophrenia Bulletin 40, 181191.Google Scholar
Sripada, RK, Marx, CE, King, AP, Rampton, JC, Ho, SS and Liberzon, I (2013) Allopregnanolone elevations following pregnenolone administration are associated with enhanced activation of emotion regulation neurocircuits. Biological Psychiatry 73, 10451053.Google Scholar
Tajik-Esmaeeli, S, Moazen-Zadeh, E, Abbasi, N, Shariat, SV, Rezaei, F, Salehi, B and Akhondzadeh, S (2017) Simvastatin adjunct therapy for negative symptoms of schizophrenia: a randomized double-blind placebo-controlled trial. International Clinical Psychopharmacology 32, 8794.Google Scholar
Takao, K, Kobayashi, K, Hagihara, H, Ohira, K, Shoji, H, Hattori, S, Koshimizu, H, Umemori, J, Toyama, K, Nakamura, HK, Kuroiwa, M, Maeda, J, Atsuzawa, K, Esaki, K, Yamaguchi, S, Furuya, S, Takagi, T, Walton, NM, Hayashi, N, Suzuki, H, Higuchi, M, Usuda, N, Suhara, T, Nishi, A, Matsumoto, M, Ishii, S and Miyakawa, T (2013) Deficiency of schnurri-2, an MHC enhancer binding protein, induces mild chronic inflammation in the brain and confers molecular, neuronal, and behavioral phenotypes related to schizophrenia. Neuropsychopharmacology 38, 14091425.Google Scholar
Tousi, B (2015) The emerging role of bexarotene in the treatment of Alzheimer's disease: current evidence. Neuropsychiatric Disease and Treatment 11, 311315.Google Scholar
Upthegrove, R, Manzanares-Teson, N and Barnes, NM (2014) Cytokine function in medication-naive first episode psychosis: a systematic review and meta-analysis. Schizophrenia Research 155, 101108.Google Scholar
Usall, J, Huerta-Ramos, E, Labad, J, Cobo, J, Nunez, C, Creus, M, Pares, GG, Cuadras, D, Franco, J, Miquel, E, Reyes, JC and Roca, M (2016) Raloxifene as an adjunctive treatment for postmenopausal women with schizophrenia: a 24-week double-blind, randomized, parallel, placebo-controlled trial. Schizophrenia Bulletin 42, 309317.Google Scholar
van Beveren, NJ, Schwarz, E, Noll, R, Guest, PC, Meijer, C, de Haan, L and Bahn, S (2014) Evidence for disturbed insulin and growth hormone signaling as potential risk factors in the development of schizophrenia. Translational Psychiatry 4, e430.Google Scholar
Vane, JR, Bakhle, YS and Botting, RM (1998) Cyclooxygenases 1 and 2. Annual Review of Pharmacology and Toxicology 38, 97120.Google Scholar
Vasovic, V, Banic, B, Jakovljevic, V, Tomic, Z and Milic-Djordjevic, V (2008) Effect of aminophylline on aspirin penetration into the central nervous system in rats. European Journal of Drug Metabolism and Pharmacokinetics 33, 2330.Google Scholar
Vincenzi, B, Stock, S, Borba, CP, Cleary, SM, Oppenheim, CE, Petruzzi, LJ, Fan, X, Copeland, PM, Freudenreich, O, Cather, C and Henderson, DC (2014) A randomized placebo-controlled pilot study of pravastatin as an adjunctive therapy in schizophrenia patients: effect on inflammation, psychopathology, cognition and lipid metabolism. Schizophrenia Research 159, 395403.Google Scholar
Wakade, CG, Mahadik, SP, Waller, JL and Chiu, FC (2002). Atypical neuroleptics stimulate neurogenesis in adult rat brain. Journal of Neuroscience Research 69, 7279.Google Scholar
Watabe, M, Kato, TA, Monji, A, Horikawa, H and Kanba, S (2012) Does minocycline, an antibiotic with inhibitory effects on microglial activation, sharpen a sense of trust in social interaction? Psychopharmacology 220, 551557.Google Scholar
Weiser, M, Burshtein, S and Fodoreanu, L (2012) A randomized trial administering aspirin, minocycline or pramipexole vs placebo as add-on to antipsychotics in patients with schizophrenia or schizoaffective disorder [abstract]. Neuropsychopharmacology 38, S314S446.Google Scholar
Weiser, M, Levi, L, Burshtein, S, Hagin, M, Matei, VP, Podea, D, Miclutia, I, Tiugan, A, Pacala, B, Grecu, IG, Noy, A, Zamora, D and Davis, JM (2017) Raloxifene plus antipsychotics versus placebo plus antipsychotics in severely Ill decompensated postmenopausal women with schizophrenia or schizoaffective disorder: a randomized controlled trial. Journal of Clinical Psychiatry 78, e758e765.Google Scholar
Weiser, M, Levi, L, Burshtein, S, Chirita, R, Cirjaliu, D, Gonen, I, Yolken, R, Davidson, M, Zamora, D and Davis, JM (2019) The effect of minocycline on symptoms in schizophrenia: results from a randomized controlled trial. Schizophrenia Research 206, 325332.Google Scholar
Yin, C, Ackermann, S, Ma, Z, Mohanta, SK, Zhang, C, Li, Y, Nietzsche, S, Westermann, M, Peng, L, Hu, D, Bontha, SV, Srikakulapu, P, Beer, M, Megens, RTA, Steffens, S, Hildner, M, Halder, LD, Eckstein, HH, Pelisek, J, Herms, J, Roeber, S, Arzberger, T, Borodovsky, A, Habenicht, L, Binder, CJ, Weber, C, Zipfel, PF, Skerka, C and Habenicht, AJR (2019) Apoe attenuates unresolvable inflammation by complex formation with activated C1q. Nature Medicine 25, 496506.Google Scholar
Yrjanheikki, J, Keinanen, R, Pellikka, M, Hokfelt, T and Koistinaho, J (1998) Tetracyclines inhibit microglial activation and are neuroprotective in global brain ischemia. Proceedings of the National Academy of Sciences of the United States of America 95, 1576915774.Google Scholar
Zhang, W, Wang, T, Qin, L, Gao, HM, Wilson, B, Ali, SF, Zhang, W, Hong, JS and Liu, B (2004) Neuroprotective effect of dextromethorphan in the MPTP Parkinson's disease model: role of NADPH oxidase. FASEB Journal 18, 589591.Google Scholar
Zhang, J-h, Chen, B and Jian-rui, L (2015) Treatment effect of risperidone alone and combined with N-acetly-cysteine for first-episode schizophrenic patients [Chinese article]. Journal of Clinical Psychiatry 25, 394396.Google Scholar
Zhang, L, Zheng, H, Wu, R, Zhu, F, Kosten, TR, Zhang, XY and Zhao, J (2018) Minocycline adjunctive treatment to risperidone for negative symptoms in schizophrenia: association with pro-inflammatory cytokine levels. Progress in Neuro-Psychopharmacology and Biological Psychiatry 85, 6976.Google Scholar
Zheng, W, Cai, DB, Yang, XH, Ungvari, GS, Ng, CH, Muller, N, Ning, YP and Xiang, YT (2017) Adjunctive celecoxib for schizophrenia: a meta-analysis of randomized, double-blind, placebo-controlled trials. Journal of Psychiatric Research 92, 139146.Google Scholar
Figure 0

Table 1. Main types of medication with anti-inflammatory actions

Figure 1

Fig. 1. PRISMA flow diagram of the performed literature search.

Figure 2

Fig. 2. Forest Plot Showing Effect Sizes for Anti-Inflammatory Therapies in Schizophrenia.

Supplementary material: File

Çakici et al. supplementary material

Çakici et al. supplementary material
Download Çakici et al. supplementary material(File)
File 444.6 KB