Hostname: page-component-8448b6f56d-c47g7 Total loading time: 0 Render date: 2024-04-23T23:01:56.668Z Has data issue: false hasContentIssue false

Systematic review and meta-analysis of the efficacy and safety of minocycline in schizophrenia

Published online by Cambridge University Press:  09 February 2017

Marco Solmi*
Affiliation:
Department of Neurosciences, University of Padova, Padova, Italy Local Health Unit 17, ULSS 17, Mental Health Department, Padova, Italy Institute for Clinical Research and Education in Medicine (IREM), Padova, Italy
Nicola Veronese
Affiliation:
Institute for Clinical Research and Education in Medicine (IREM), Padova, Italy Department of Medicine (DIMED), Geriatrics Section, University of Padova, Padova, Italy
Nita Thapa
Affiliation:
Kaski Sewa Hospital and Research Centre, Pokhara, Nepal
Silvia Facchini
Affiliation:
Department of Medicine (DIMED), Geriatrics Section, University of Padova, Padova, Italy
Brendon Stubbs
Affiliation:
Physiotherapy Department, South London and Maudsley NHS Foundation Trust, London, United Kingdom Health Service and Population Research Department, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
Michele Fornaro
Affiliation:
New York Psychiatric Institute, Columbia University, New York, New York, USA
André F. Carvalho
Affiliation:
Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
Christoph U. Correll
Affiliation:
The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, New York, USA Hofstra Northwell School of Medicine, Hempstead, New York, USA The Feinstein Institute for Medical Research, Manhasset, New York, USA Albert Einstein College of Medicine, Bronx, New York, USA
*
*Address correspondence to: Marco Solmi, Department of Neurosciences, University of Padua, Via Giustiniani, 2, 35128 Padova, Italy. (Email: marco.solmi83@gmail.com)
Rights & Permissions [Opens in a new window]

Abstract

Objective

Our aim was to perform an updated systematic review and meta-analysis on the efficacy and safety of adjunctive minocycline as a treatment of schizophrenia.

Methods

We conducted a PubMed/Scopus database search from inception to 3 February 2016 for randomized, placebo-controlled trials (RCTs), open non-randomized studies, and case reports/series evaluating minocycline in patients with schizophrenia. Random-effects meta-analysis of positive, negative, depressive, and cognitive symptom rating scales, discontinuation and adverse effects rates calculating standardized mean difference (SMD), and risk ratios±95% confidence intervals (CI95%) were calculated.

Results

Six RCTs were eligible (minocycline n=215, placebo n=198) that demonstrated minocycline’s superiority versus placebo for reducing endpoint Positive and Negative Syndrome Scale (PANSS) total scores (SMD=–0.59; CI95%=[1.15, –0.03]; p=0.04), negative (SMD=–0.76; CI95%=[–1.21, –0.31]; p=0.001); general subscale scores (SMD=–0.44; CI95%=[–0.88, –0.00]; p=0.05), Clinical Global Impressions scores (SMD=–0.50; CI95%=[–0.78, –0.22]; p<0.001); and executive functioning (SMD=0.22; CI95%=[0.01, 0.44]; p=0.04). Endpoint PANSS positive symptom scores (p=0.13), depression rating scale scores (p=0.43), attention (p=0.47), memory (p=0.52), and motor speed processing (p=0.50) did not significantly differ from placebo, before execution of a trim-and-fill procedure. Minocycline did not differ compared to placebo on all-cause discontinuation (p=0.56), discontinuation due to inefficacy (p=0.99), and intolerability (p=0.51), and due to death (p=0.32). Data from one open-label study (N=22) and three case series (N=6) were consistent with the metaanalytic results.

Conclusions

Minocycline appears to be an effective adjunctive treatment option in schizophrenia, improving multiple relevant disease dimensions. Moreover, minocycline has an acceptable safety and tolerability profile. However, more methodologically sound and larger RCTs remain necessary to confirm and extend these results.

Type
Review Articles
Copyright
© Cambridge University Press 2017 

Introduction

Minocycline, a second-generation tetracycline antibiotic, has pleiotropic mechanisms of action in the central nervous system that include the modulation of glutamate-N-methyl-d-aspartate receptors,Reference Chaves, Marque and Trzesniak 1 Reference Macdonald, Kelly, Allen, Noble and Kanegis 6 a decrease in oxidative and nitrosative stress (O&NS),Reference Monte, de Souza and McIntyre 2 , Reference Hanson, Healey, Wolf and Kohler 7 as well as putative anti-inflammatory effects (e.g., a decrease in the production of tumor necrosis factor-α and interferon-γ by activated microglia).Reference Liaury, Miyaoka and Tsumori 8 , Reference Seki, Kato and Monji 9 Minocycline has demonstrated neuroprotective properties in such neurodegenerative disorders as Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis (ALS),Reference Hashimoto 3 , Reference Hashimoto, Ishima, Fujita and Zhang 4 , Reference Zhang, Narayanan and Friedlander 10 in addition to ischemia.Reference Yrjanheikki, Tikka, Keinanen, Goldsteins, Chan and Koistinaho 11

Beyond dopaminergic signaling dysfunction, inflammatory,Reference Davis, Moylan, Harvey, Maes and Berk 12 , Reference Muller, Weidinger, Leitner and Schwarz 13 glutamatergic,Reference Hu, MacDonald, Elswick and Sweet 14 , Reference Zink and Correll 15 and oxidative stress pathwaysReference Reus, Fries and Stertz 16 , Reference Shim, Shuman and Duncan 17 may be involved in the pathophysiology of schizophrenia, particularly in relation to negative and cognitive symptoms.Reference Fillman, Weickert and Lenroot 18 Reference Tuominen, Tiihonen and Wahlbeck 20 In addition, these pathways may interact to drive neuroprogression in this illness.Reference Davis, Moylan, Harvey, Maes and Berk 12 NegativeReference Ventura, Subotnik and Gitlin 21 Reference Galderisi, Bucci and Mucci 24 and cognitiveReference Agid, Siu, Pappadopulos, Vanderburg and Remington 25 , Reference Hofer, Baumgartner and Bodner 26 symptoms of schizophrenia are the main determinants of the prognosis and course of schizophrenia.Reference Remberk, Bazynska and Bronowska 22

The mechanism of action of minocycline in schizophrenia has been reviewed elsewhere.Reference Zhang and Zhao 27 This second-generation tetracycline antibiotic has anti-inflammatory properties, inhibits microglial activation, decreases O&NS, inhibits apoptosis, and modulates glutamate-mediated excitotoxicity. Minocycline may have beneficial effects in patients with schizophrenia in whom antipsychotic agents are insufficiently effective on neuroinflammation involving microglia,Reference Monji, Kato and Mizoguchi 28 apoptotic mechanisms,Reference Glantz, Gilmore, Lieberman and Jarskog 29 oxidative stress.Reference Sertan Copoglu, Virit and Hanifi Kokacya 30 and glutamate dysfunction,Reference Howes, McCutcheon and Stone 31 which appear to interact with dopamine- and serotonin-related signaling, thus promoting neuroprogression of this severe mental illness.Reference Davis, Moylan, Harvey, Maes and Berk 12 While second-generation antipsychotics (SGAs) mainly act on positive symptoms modulating dopamine and serotonin pathways with mostly questionable or minor effects on glutamate signaling,Reference Xu, Gullapalli and Frost 32 Reference Koprivica, Regardie and Wolff 34 the minocycline pharmacodynamic profile may be used in a multimodal treatment approach in schizophrenia. In this context, add-on minocycline may mechanistically address the areas that are mainly not improved by available antipsychotics (namely, improve negative and cognitive symptoms) and which remain a clear unmet need in the therapeutic management of schizophrenia.Reference Millan, Fone, Steckler and Horan 35 , Reference Millan, Agid and Brune 36

Thus, minocycline has been investigated as a novel therapeutic target for schizophrenia. Two previous meta-analyses have investigated the effects of minocycline on the symptom domains of schizophrenia.Reference Oya, Kishi and Iwata 37 , Reference Iwata, Nakajima and Suzuki 38 Overall, these studies have concluded that minocycline is superior to placebo in improving total, negative, and general symptom scores on the Positive and Negative Syndrome Scale (PANSS),Reference Kay, Fiszbein and Opler 39 the Scale for the Assessment of Negative Symptoms (SANS),Reference Andreasen 40 and the Clinical Global Impressions–Severity scale (CGI–S),Reference Guy 41 whereas no significant differences relative to placebo were found for positive and depressive symptoms and global cognitive symptoms, but minocycline did appear to be safe and tolerable.Reference Iwata, Nakajima and Suzuki 38 Those conclusions deserve reassessment, as both meta-analyses were preliminary and based on four and two studies, with 330 and 100 patients,Reference Oya, Kishi and Iwata 37 , Reference Iwata, Nakajima and Suzuki 38 and as additional studies have become available.Reference Ghanizadeh, Dehbozorgi, OmraniSigaroodi and Rezaei 42 , Reference Kelly, Sullivan and McEvoy 43 Therefore, a larger sample size will increase the power and confidence in the findings, potentially enabling meaningful subgroup or meta-regression analyses to identify potential sources of heterogeneity.

We aimed at providing a wide overview of the extant literature on minocycline’s role in the treatment of schizophrenia, consisting of a descriptive plus a systematic review not limited to randomized controlled trials (RCTs). We further aimed to reassess minocycline’s efficacy and safety in a formal meta-analysis, with a larger sample size, focusing on psychopathology and cognition as well as tolerability and safety.

Methods

This systematic review adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) Statement,Reference Moher, Liberati, Tetzlaff and Altman 44 following a predetermined, but unpublished, protocol.

Search strategy

An electronic literature search was conducted in PubMed and Scopus from database inception until 3 February 2016 by two independent reviewers (M.S, N.V.), using the search terms (minocycline) AND (“schizophrenia” OR “psychosis” OR “psychotic disorder” OR “schizoaffective”) to identify RCTs, open-label trials, and case series or reports that investigated the efficacy and safety of minocycline in patients diagnosed with schizophrenia or schizoaffective disorder.

Inclusion and exclusion criteria

Studies eligible for the meta-analysis were RCTs that (1) compared minocycline with placebo; (2) included patients diagnosed with schizophrenia or schizoaffective disorder according to structured clinical assessments; and (3) reported efficacy data using a standardized rating scale, such as the Scale for Assessment of Negative Symptoms (SANS),Reference Andreasen 40 the Positive and Negative Syndrome Scale (PANSS),Reference Kay, Fiszbein and Opler 39 the Brief Psychiatric Rating Scale (BPRS),Reference Overall and Gorham 45 the Clinical Global Impressions Scale (CGI),Reference Guy 41 the Calgary Depression Scale for Schizophrenia (CDSS),Reference Lancon, Auquier, Reine, Bernard and Toumi 46 the Hamilton Depression Rating Scale (HDRS),Reference Hamilton 47 the Beck Depression Inventory (BDI),Reference Beck and Alford 48 the MATRICS Consensus Cognitive Battery (MCCB),Reference Nuechterlein, Green and Kern 49 , Reference Green and Nuechterlein 50 the Cambridge Neuropsychological Test Automated Battery, 51 discontinuation rates, frequencies of side effects, results on extrapyramidal symptom scales (such as the Extrapyramidal Symptom Rating ScaleReference Monte, de Souza and McIntyre 2 and the Abnormal Involuntary Movement ScaleReference Guy 41 ), as well as weight change and metabolic abnormalities. For the systematic review, we also included case reports, case series, and open-label studies, reporting the effects of the use of minocycline in patients affected by schizophrenia or schizoaffective disorder. Studies were excluded if they reported on minocycline for patients with a different disease or were on a different drug.

Outcomes

The primary outcome was the PANSS total endpoint score. Secondary outcomes included PANSS positive, negative, and general endpoint subscores; SANS and CGI scores; depressive rating scales endpoint scores; cognitive endpoint scores; and all-cause and specific cause discontinuation rates. Safety outcomes included extrapyramidal symptom scales and individual side-effect frequencies. When studies reported cognitive outcomes, we grouped the individual tests into broad domains to enable pooled analyses across different tests (for details, see Supplementary Table 1).

Data extraction

Three reviewers (M.S., N.V., S.F.) independently extracted data from the included studies into a standardized Microsoft Excel spreadsheet. Any disagreement was resolved by consensus. The following information was extracted: author, year, country, study design, sponsor/funding, inclusion and exclusion criteria, trial duration, setting, sample size, population demographics, minocycline and other medication doses, outcome measures, baseline, follow-up, and change in all rating scales, discontinuation rates, side effects, and quality indicators. Whenever data were not reported or we needed clarification, we contacted authors up to three times requesting additional information.

Quality assessment

Evaluation of methodological study quality was conducted by two independent reviewers (M.S., N.V.) using the Cochrane Collaboration’s tool for assessing risk of bias.Reference Higgins, Altman and Gotzsche 53 This tool includes six domains that can indicate low, unclear, or high risk of bias. Considering the six domains, a study is defined as having low risk of bias when all domains indicate low risk of bias, unclear risk of bias when one or more domains indicate unclear risk of bias, and high risk of bias when high risk of bias is present for one or more key domains.

Data analysis

The meta-analysis was performed using Review Manager 54 (v. 5.1 for Windows) (http://tech.cochrane.org/revman). All outcomes were meta-analyzed when at least two studies provided data for a given outcome. When combining studies, the random effects modelReference DerSimonian and Kacker 55 , Reference DerSimonian and Laird 56 was used to account for study heterogeneity. For continuous data, we calculated the standardized mean difference (SMD) with its 95% confidence interval as the effect size; for dichotomous data, we used risk ratio (RR) with its 95% confidence interval. The SMD as effect size allowed us to group together different scales measuring the same dimensions (e.g., depression). Study heterogeneity was measured using χReference Monte, de Souza and McIntyre 2 and I 2 statistics, with p<0.05 for χ2 and ≥50% for I 2 indicating significant heterogeneity.Reference Higgins, Thompson, Deeks and Altman 57 We compared endpoint rating scale values, all-cause and specific-cause discontinuation, and side-effect rates. When heterogeneity was high, as defined by I 2≥50%, when at least four studies were available, meta-regression analyses were performed with Comprehensive Meta-Analysis (v. 3), 58 investigating the following potential moderator variables: age, sex, study duration, and illness duration.

Finally, funnel plots were visually inspected, and Egger’s testReference Egger, Davey Smith, Schneider and Minder 59 and Begg–Mazumdar Kendall’s tauReference Begg and Mazumdar 60 were utilized to determine if a publication bias was likely, and if it was part of the trim-and-fill procedure,Reference Duval and Tweedie 61 it was run in order to evaluate if the results changed after imputing potentially missing studies.

Results

Search results

The study selection flow is depicted in Figure 1. Out of 322 initial hits, 307 were excluded through title/abstract reading. A total of 15 full texts were reviewed, and 1 study was excluded since it reported neuroimaging data on a previously reported sample,Reference Chaves, Marque and Maia-de-Oliveira 62 2 because they were meta-analyses,Reference Oya, Kishi and Iwata 37 , Reference Iwata, Nakajima and Suzuki 38 and 2 were trial protocols.Reference Lisiecka, Suckling and Barnes 63 , Reference Fekadu, Mesfin and Medhin 64 Among the remaining 10 studies, 6 RCTs were included in the quantitative meta-analysis.Reference Ghanizadeh, Dehbozorgi, OmraniSigaroodi and Rezaei 42 , Reference Kelly, Sullivan and McEvoy 43 , Reference Chaudhry, Hallak and Husain 65 Reference Levkovitz, Mendlovich and Riwkes 68 Out of the four studies included in the systematic review, one was an open-label study Reference Miyaoka, Yasukawa, Yasuda, Hayashida, Inagaki and Horiguchi 69 and three were case series.Reference Kelly, Vyas and Richardson 70 Reference Qurashi, Collins, Chaudhry and Husain 72

Figure 1 PRISMA flowchart.

Included studies, treatments, and participants (Table 1)

We meta-analyzed 6 placebo controlled RCTs,Reference Ghanizadeh, Dehbozorgi, OmraniSigaroodi and Rezaei 42 , Reference Kelly, Sullivan and McEvoy 43 , Reference Chaudhry, Hallak and Husain 65 Reference Levkovitz, Mendlovich and Riwkes 68 including 215 patients taking minocycline and 198 patients taking placebo. In the minocycline group, the patients were on average 29.91±10.2 years old, their age of illness onset was 20.24±5.28 years, illness duration was 17.79±12.98 years, duration of education was 10.39±3.6 years, and 67.92 % were male. In the placebo group, patients were 29.88±9.9 years of age, their age of illness onset was 20.3±5.04 years, illness duration was 20.18±14.69 years, duration of education was 10.21±3.79 years, and 73.4% were male. The mean study duration was 19.7 (range=8–24) weeks, and all 6 studies used minocycline (target dose=200 mg/day) as an augmentation strategy. Baseline antipsychotics included risperidone in three studies,Reference Ghanizadeh, Dehbozorgi, OmraniSigaroodi and Rezaei 42 , Reference Khodaie-Ardakani, Mirshafiee and Farokhnia 66 , Reference Liu, Guo and Wu 67 clozapine in one study,Reference Kelly, Sullivan and McEvoy 43 and mixed antipsychotics in two studies.Reference Chaudhry, Hallak and Husain 65 , Reference Levkovitz, Mendlovich and Riwkes 68 Two studies were conducted in Iran,Reference Ghanizadeh, Dehbozorgi, OmraniSigaroodi and Rezaei 42 , Reference Khodaie-Ardakani, Mirshafiee and Farokhnia 66 and one each in the United States,Reference Kelly, Sullivan and McEvoy 43 China,Reference Liu, Guo and Wu 67 Brazil and Pakistan,Reference Chaudhry, Hallak and Husain 65 and Israel.Reference Levkovitz, Mendlovich and Riwkes 68 Two studies allowed inclusion of patients with schizoaffective disorder.Reference Kelly, Sullivan and McEvoy 43 , Reference Chaudhry, Hallak and Husain 65 All studies except oneReference Kelly, Sullivan and McEvoy 43 used the PANSS, four studies used the CGI,Reference Kelly, Sullivan and McEvoy 43 , Reference Chaudhry, Hallak and Husain 65 , Reference Liu, Guo and Wu 67 , Reference Levkovitz, Mendlovich and Riwkes 68 four studies used the SANS,Reference Ghanizadeh, Dehbozorgi, OmraniSigaroodi and Rezaei 42 , Reference Kelly, Sullivan and McEvoy 43 , Reference Liu, Guo and Wu 67 , Reference Levkovitz, Mendlovich and Riwkes 68 four used rating scales for depression,Reference Ghanizadeh, Dehbozorgi, OmraniSigaroodi and Rezaei 42 , Reference Kelly, Sullivan and McEvoy 43 , Reference Khodaie-Ardakani, Mirshafiee and Farokhnia 66 , Reference Levkovitz, Mendlovich and Riwkes 68 and three studies assessed cognitive functioning.Reference Kelly, Sullivan and McEvoy 43 , Reference Liu, Guo and Wu 67 , Reference Levkovitz, Mendlovich and Riwkes 68

Table 1 Study, patient, illness and treatment characteristics on the meta-analyzed studies

BPRS=Brief Psychiatric Rating Scale; CDRS=Calgary Depression Rating Scale; CRP=C-reactive protein; DB=double blind; mcy=minocycline; NOS=not otherwise specified; OL=open-label; PC=placebo-controlled; PLC=placebo; R=randomized; SANS=Scale for Assessment of Negative Symptoms; SGA=second-generation antipsychotic.

The open-label study conducted in JapanReference Miyaoka, Yasukawa, Yasuda, Hayashida, Inagaki and Horiguchi 69 lasted 4 weeks and included 22 patients with schizophrenia, with a mean age of 31.2±5.5 years, mean age of illness onset of 22.8±9.73 years, and illness duration of 3.4±2.3 years, with 63.6% being male.

Three case series—one from the United Kingdom,Reference Qurashi, Collins, Chaudhry and Husain 72 one from India,Reference Jhamnani, Shivakumar, Kalmady, Rao and Venkatasubramanian 71 and one from the United StatesReference Kelly, Vyas and Richardson 70 —with two cases in each report described changes on several rating scales in patients with schizophrenia.

Quality assessment (Supplementary Table2) or randomized placebo controlled studies

According to Cochrane Collaboration’s tool for assessing risk of bias,Reference Higgins, Altman and Gotzsche 53 two studies had an unclear risk of bias,Reference Ghanizadeh, Dehbozorgi, OmraniSigaroodi and Rezaei 42 , Reference Levkovitz, Mendlovich and Riwkes 68 while each of the others had a low risk of bias.

Meta-analysis: efficacy

All primary and secondary outcome results are provided in Table 2. The minocycline group had lower endpoint scores compared to placebo in PANSS total score (SMD=–0.59; CI 95%=[–1.15, –0.03]; p=0.04); PANSS negative score (SMD=–0.76; CI 95%=[–1.21, –0.31]; p=0.001); SANS score (SMD=0.60; CI 95%=[–0.94, –0.27]; p<0.001); PANSS general score (SMD=–0.44; CI 95%=[–0.88, –0.00]; p=0.05); CGI–S (SMD=–0.50; CI 95%=[–0.78, –0.22]; p<0.001) and higher (better) executive functioning scores (SMD=0.22; CI 95%=[0.01, 0.44]; p=0.04). Results were significantly heterogeneous for the three PANSS-based findings, but not for the remainder of the outcomes that favored minocycline (Table 2).

Table 2 Meta-analysis and publication bias of efficacy and cognitive outcomes

CGI=Clinical Global Impression Scale; PANSS=Positive and Negative Syndrome Scale; SANS=Scale for Assessment of Negative Symptoms; SMD=standardized mean difference.

Depressive scores include data from the Calgary Depression Scale, the Hamilton Depression Rating Scale, and the Beck Depression Inventory. Significant results in bold.

Endpoint PANSS positive symptom scores (p=0.13), depression rating scale scores (p=0.43), attention (p=0.47), memory (p=0.52), and motor speed processing (p=0.50) did not significantly differ from placebo before the trim-and-fill procedure. These nonsignificant findings were not significantly heterogeneous, with the exception of the results for motor speed and memory (Table 2). All-cause discontinuation (p=0.56), discontinuation due to inefficacy (p=0.99), discontinuation due to intolerability (p=0.51), and discontinuation due to death (p=0.32) did not differ between the minocycline and placebo groups.

Meta-analysis: publication bias and trim-and-fill (Table 2)

Publication bias test and trim-and-fill procedures did not show any bias in our results. However, the failsafe number was one, suggesting a weak consistency of this result.

Meta-analysis: meta-regression Analyses (Table 3)

No significant moderators of primary and secondary outcomes with at least four studies contributing data emerged, including baseline values of each rating scale, country of the study (Asia vs. others), trial duration, baseline antipsychotic (risperidone vs. others), and difference of mean age between the minocycline and placebo groups.

Table 3 Meta-regression analysis of the heterogeneous findings

CGI=Clinical Global Impression Scale; PANSS=Positive and Negative Syndrome Scale.

Meta-analysis: safety and tolerability

No significant difference emerged between minocycline and placebo as concerns suicide (studies n=2, p=0.79),Reference Chaudhry, Hallak and Husain 65 , Reference Levkovitz, Mendlovich and Riwkes 68 pigmentation (studies n=2, p=0.53),Reference Chaudhry, Hallak and Husain 65 , Reference Levkovitz, Mendlovich and Riwkes 68 loss of appetite (studies n=3, p=0.99),Reference Kelly, Sullivan and McEvoy 43 , Reference Chaudhry, Hallak and Husain 65 , Reference Liu, Guo and Wu 67 dizziness (studies n=3, p=0.55),Reference Kelly, Sullivan and McEvoy 43 , Reference Chaudhry, Hallak and Husain 65 , Reference Liu, Guo and Wu 67 vomiting (studies n=2, p=0.43),Reference Kelly, Sullivan and McEvoy 43 , Reference Chaudhry, Hallak and Husain 65 nausea (studies n=3, p=0.70),Reference Kelly, Sullivan and McEvoy 43 , Reference Chaudhry, Hallak and Husain 65 , Reference Liu, Guo and Wu 67 extrapyramidal symptoms both as reported by investigators (studies n=3, p=0.95)Reference Kelly, Sullivan and McEvoy 43 , Reference Chaudhry, Hallak and Husain 65 , Reference Liu, Guo and Wu 67 and measured with the Extrapyramidal Symptoms Rating ScaleReference Chouinard and Margolese 52 (studies n=2, p=0.72),Reference Khodaie-Ardakani, Mirshafiee and Farokhnia 66 , Reference Levkovitz, Mendlovich and Riwkes 68 constipation (studies n=3, p=0.68),Reference Kelly, Sullivan and McEvoy 43 , Reference Liu, Guo and Wu 67 , Reference Levkovitz, Mendlovich and Riwkes 68 and dry mouth (studies n=2, p=0.56)Reference Kelly, Sullivan and McEvoy 43 , Reference Liu, Guo and Wu 67 . However, headache was significantly more frequent in the placebo group (studies n=2, p=0.01).Reference Kelly, Sullivan and McEvoy 43 , Reference Chaudhry, Hallak and Husain 65

Systematic review: efficacy and safety

One open-label studyReference Miyaoka, Yasukawa, Yasuda, Hayashida, Inagaki and Horiguchi 69 that included 22 patients affected by schizophrenia resistant to other standard treatments reported that minocycline reduced PANSS positive scores to 40.4% at 8 weeks, PANSS negative scores to 44%, and PANSS general scores to 52.1%. The three case series described improvements in BPRS scoresReference Overall and Gorham 45 in two patients with paranoid schizophrenia,Reference Qurashi, Collins, Chaudhry and Husain 72 improvements in SANS scores and C-reactive protein values in one patient with undifferentiated schizophrenia and one with paranoid schizophrenia, both with high C-reactive protein,Reference Jhamnani, Shivakumar, Kalmady, Rao and Venkatasubramanian 71 and improvements in BPRS and SANS scores plus CDSS in one case out of two patients affected by catatonic schizophrenia, who wanted to continue taking minocycline despite mild nausea, constipation, and abdominal pain.Reference Kelly, Vyas and Richardson 70 In three of these cases, patients had a history of failing to respond to clozapine.

Discussion

The results of this, to date largest, systematic review and meta-analysis of the randomized controlled evidence of the efficacy and safety of minocycline for the treatment of schizophrenia suggests a significant beneficial effect of minocycline on several psychopathological and cognitive domains in schizophrenia. Effect sizes were small for the one positive effect on cognition and in the medium range for the significant psychopathology improvements, with a near-large effect size for the PANSS-based negative symptom improvement. The lack of any effect on depression or extrapyramidal symptoms (EPS) ratings strengthens the results regarding improved negative symptoms, which has long been an elusive goal in schizophrenia, as depression and EPS can impose as secondary negative symptoms.Reference Carbon and Correll 73 We provide a novel insight into the cognitive effects of minocycline, which are in contrast with a former meta-analysisReference Iwata, Nakajima and Suzuki 38 that suggested an effect of minocycline on attention/vigilance in schizophrenia. Notwithstanding the fact that our analyses did not confirm a role of minocycline in improving attention in schizophrenia, we report an improvement in executive functioning. Furthermore, results from a former meta-analysisReference Oya, Kishi and Iwata 37 that indicated beneficial effects of minocycline compared to placebo on the psychopathological domains of schizophrenia—including efficacy on PANSS total, the negative and general subscales, in SANS score, and CGI–S scores—are now confirmed after adding two more trials (+50%)Reference Ghanizadeh, Dehbozorgi, OmraniSigaroodi and Rezaei 42 , Reference Kelly, Sullivan and McEvoy 43 with 83 patients (+25%). However, we suggest that at this stage more well-designed RCTs are necessary to further investigate the effects of minocycline on the positive symptoms of schizophrenia.

Since minocycline has a different pharmacodynamic profile than SGAs and complementary clinical targets, all included trials utilized an “add-on” design, adding minocycline to the SGAs that had yielded insufficient results. Similar to major depressive disorder, where anti-inflammatory agents have been added to antidepressants aimed at improving cognition,Reference Carvalho, Miskowiak and Hyphantis 74 a multimodal approach may also help to address unmet treatment needs in schizophrenia.

Beyond the minocycline efficacy data, its safety and tolerability profile, alongside the increased subjective well-being reported in case reports, suggest that minocycline may also facilitate pharmacological compliance, which indeed is often an object of concern in patients with schizophrenia.Reference Kane, Kishimoto and Correll 75 However, minocycline has also been associated with triggering or worsening severe autoimmune conditions, such as systemic lupus erythematosus, autoimmune hepatitis, hyperthyroidism, neutropenia, and polyarthritis nodosa;Reference Tehrani, Nash-Goelitz, Adams, Dahiya and Eilers 76 Reference Ahmed, Kelsey and Shariff 79 hence, patients treated with minocycline should be carefully monitored.

The present work has several strengths. The sample size increased from 173 patients with schizophrenia on minocycline and 157 on placeboReference Oya, Kishi and Iwata 37 to 215 and 198, respectively. Then, we also retested previous evidence controlling for publication bias and potential moderators, suggesting the need for more studies assessing minocycline’s efficacy for positive symptoms of schizophrenia. Moreover, even if our analyses did not suggest any significant moderator of the observed effect sizes, they still conferred more solid and methodologically sound evidence. In addition, our results describe a role for minocycline in enhancing executive functioning in schizophrenia, a core feature of this enduring disease. Finally, we added a descriptive and systematic review of nonrandomized literature, providing further support to our and other colleagues’ conclusions,Reference Oya, Kishi and Iwata 37 consisting of an open-label studyReference Miyaoka, Yasukawa, Yasuda, Hayashida, Inagaki and Horiguchi 69 and several case series.Reference Kelly, Vyas and Richardson 70 Reference Qurashi, Collins, Chaudhry and Husain 72 Even if these latter reports do not contribute to the evidence as RCTs do, they do provide a valuable contribution in terms of clinical and real-world-based experience.

However, several factors should be considered when interpreting these results. First, although we increased the number of studies and patients considerably, the number of trials and randomized patients is still quite modest. Thus, although the results are more robust than before and although there does not appear to be a relevant publication bias, additional and larger studies with minocycline and with mechanistically similar molecules are needed. Second, due to the still small evidence base, our subgroup and meta-regression had to remain exploratory. Since several relevant outcomes had a significant heterogeneity of findings, a larger database will be needed to help identify subgroups of patients, and to design features or treatment characteristics that increase the likelihood of benefiting from minocycline. Third, since only one study had clozapine as the baseline antipsychotic, it is unclear from the current data if failure to sufficiently improve to a non-clozapine antipsychotic or to clozapine would yield different outcomes with minocycline augmentation. Fourth, since all RCTs targeted 200 mg of minocycline per day, data are lacking regarding potential dose–response relationships. Finally, studies of add-on minocycline did not assess all relevant cognitive domains. For our analysis of changes in cognitive domain scores, we pooled different cognitive tests assessing similar cognitive domains. Even if this could be considered an approach that accounts for the heterogeneity of cognitive domains definitions, it could be argued that the results have reduced specificity.

Conclusions

In conclusion, based on the currently available, modest database, minocycline appears to be an effective treatment option for patients with schizophrenia who have had insufficient benefits from antipsychotic treatment, with positive effects on global severity of illness, negative symptoms, general psychopathology, and executive functions, and possibly on positive symptoms. In addition, currently ongoing trials whose protocols have been recently publishedReference Lisiecka, Suckling and Barnes 63 , Reference Fekadu, Mesfin and Medhin 64 are hoped to add relevant evidence and provide a more detailed picture of minocycline’s clinical, functional, and cognitive effects in patients with schizophrenia. If, in fact, minocycline is a viable adjunctive treatment option for patients with schizophrenia, its various pharmacological mechanisms of actions should stimulate the development of agents that have similar or enhanced properties.

Statement of Authorship

Marco Solmi, Nicola Veronese, Silvia Facchini, and Nita Thapa conducted literature screening, data extraction, and statistical analyses. Marco Solmi, Nicola Veronese, and Christoph U. Correll ran the statistical meta-analysis. Christoph U. Correll and Marco Solmi prepared the search key and the meta-analysis design, and wrote the paper, which was reviewed and edited by André Carvalho, Nicola Veronese, Brendon Stubbs, and Michele Fornaro.

Disclosures

Marco Solmi, Nicola Veronese, Silvia Facchini, André F. Carvalho, Brendon Stubbs, Michele Fornaro, and Nita Thapa hereby state that they have nothing to disclose.

Christoph U. Correll has been a consultant and/or advisor to or has received honoraria from AbbVie, Acadia, Actavis, Actelion, Alexza, Alkermes, the American Academy of Child and Adolescent Psychiatry, Bristol-Myers Squibb, Cephalon, Eli Lilly, Genentech, Gerson Lehrman Group, IntraCellular Therapies, Lundbeck, Medavante, Medscape, Merck, the National Institute of Mental Health, Janssen/J&J, Otsuka, Pfizer, ProPhase, Reviva, Roche, Sunovion, Supermus Takeda, Teva, and Vanda. He has received grant support from BMS, the Feinstein Institute for Medical Research, Janssen/J&J, the National Institute of Mental Health, the National Alliance for Research in Schizophrenia and Depression, Otsuka, and Takeda.

Conflicts of Interest

The authors hereby declare that they have no conflicts of interest to report.

Supplementary Material

To view supplementary material for this article, please visit https://doi.org/10.1017/S1092852916000638

Footnotes

No funding was directly involved in the preparation of this paper.

References

1. Chaves, C, Marque, CR, Trzesniak, C, et al. Glutamate-N-methyl-d-aspartate receptor modulation and minocycline for the treatment of patients with schizophrenia: an update. Braz J Med Biol Res. 2009; 42(11): 10021014. https://www.ncbi.nlm.nih.gov/pubmedhealth/PMH0027593/. Accessed January 13, 2017.Google Scholar
2. Monte, AS, de Souza, GC, McIntyre, RS, et al. Prevention and reversal of ketamine-induced schizophrenia related behavior by minocycline in mice: possible involvement of antioxidant and nitrergic pathways. J Psychopharmacol. 2013; 27(11): 10321043. Epub ahead of print Sep 17. http://journals.sagepub.com/doi/pdf/10.1177/0269881113503506. Accessed January 13, 2017.CrossRefGoogle ScholarPubMed
3. Hashimoto, K. Abnormality of cerebral perfusion in the posterior cingulate gyrus of a refractory patient with schizophrenia and minocycline treatment. Prog Neuropsychopharmacol Biol Psychiatry. 2010; 34(6): 1132; author reply 1133–1134. Epub ahead of print Apr 28.Google Scholar
4. Hashimoto, K, Ishima, T, Fujita, Y, Zhang, L. Antibiotic drug minocycline: a potential therapeutic drug for methamphetamine-related disorders [in Japanese]. Nihon Arukoru Yakubutsu Igakkai Zasshi. 2013; 48(2): 118125.Google Scholar
5. Zhang, L, Shirayama, Y, Iyo, M, Hashimoto, K. Minocycline attenuates hyperlocomotion and prepulse inhibition deficits in mice after administration of the NMDA receptor antagonist dizocilpine. Neuropsychopharmacology. 2007; 32(9): 20042010. Epub ahead of print Jan 17. http://www.nature.com/npp/journal/v32/n9/pdf/1301313a.pdf. Accessed January 13, 2017.CrossRefGoogle ScholarPubMed
6. Macdonald, H, Kelly, RG, Allen, ES, Noble, JF, Kanegis, LA. Pharmacokinetic studies on minocycline in man. Clin Pharmacol Ther. 1973; 14(5): 852861.Google Scholar
7. Hanson, E, Healey, K, Wolf, D, Kohler, C. Assessment of pharmacotherapy for negative symptoms of schizophrenia. Curr Psychiatry Rep. 2010; 12(6): 563571.CrossRefGoogle ScholarPubMed
8. Liaury, K, Miyaoka, T, Tsumori, T, et al. Minocycline improves recognition memory and attenuates microglial activation in Gunn rat: a possible hyperbilirubinemia-induced animal model of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2014; 50: 184190. Epub ahead of print Jan 2.CrossRefGoogle ScholarPubMed
9. Seki, Y, Kato, TA, Monji, A, et al. Pretreatment of aripiprazole and minocycline, but not haloperidol, suppresses oligodendrocyte damage from interferon-γ-stimulated microglia in co-culture model. Schizophr Res. 2013; 151(1–3): 2028. Epub ahead of print Oct 4.CrossRefGoogle Scholar
10. Zhang, W, Narayanan, M, Friedlander, RM. Additive neuroprotective effects of minocycline with creatine in a mouse model of ALS. Ann Neurol. 2003; 53(2): 267270.Google Scholar
11. Yrjanheikki, J, Tikka, T, Keinanen, R, Goldsteins, G, Chan, PH, Koistinaho, J. A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc Natl Acad Sci U S A. 1999; 96(23): 1349613500. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC23976/. Accessed January 13, 2017.CrossRefGoogle ScholarPubMed
12. Davis, J, Moylan, S, Harvey, BH, Maes, M, Berk, M. Neuroprogression in schizophrenia: pathways underpinning clinical staging and therapeutic corollaries. Aust N Z J Psychiatry. 2014; 48(6): 512529. Epub ahead of print May 6. http://journals.sagepub.com/doi/pdf/10.1177/0004867414533012. Accessed January 13, 2017.Google Scholar
13. Muller, N, Weidinger, E, Leitner, B, Schwarz, MJ. The role of inflammation in schizophrenia. Front Neurosci. 2015; 9: 372. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4612505/. Accessed January 13, 2017.Google Scholar
14. Hu, W, MacDonald, ML, Elswick, DE, Sweet, RA. The glutamate hypothesis of schizophrenia: evidence from human brain tissue studies. Ann N Y Acad Sci. 2015; 1338: 3857. Epub ahead of print Oct 14, 2014. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4363164/. Accessed January 13, 2017.CrossRefGoogle ScholarPubMed
15. Zink, M, Correll, CU. Glutamatergic agents for schizophrenia: current evidence and perspectives. Expert Rev Clin Pharmacol. 2015; 8(3): 335352.Google Scholar
16. Reus, GZ, Fries, GR, Stertz, L, et al. The role of inflammation and microglial activation in the pathophysiology of psychiatric disorders. Neuroscience. 2015; 300: 141154. Epub ahead of print May 14.CrossRefGoogle Scholar
17. Shim, S, Shuman, M, Duncan, E. An emerging role of cGMP in the treatment of schizophrenia: a review. Schizophr Res. 2015; 170(1): 226231. Epub ahead of print Dec 22.Google Scholar
18. Fillman, SG, Weickert, TW, Lenroot, RK, et al. Elevated peripheral cytokines characterize a subgroup of people with schizophrenia displaying poor verbal fluency and reduced Broca’s area volume. Mol Psychiatry. 2015; 21(8): 10901098. Epub ahead of print Jul 21. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4960447/. Accessed January 13, 2017.Google Scholar
19. Xiu, MH, Yang, GG, Tan, YL, et al. Decreased interleukin-10 serum levels in first-episode drug-naive schizophrenia: relationship to psychopathology. Schizophr Res. 2014; 156(1): 914. Epub ahead of print Apr 22.CrossRefGoogle ScholarPubMed
20. Tuominen, HJ, Tiihonen, J, Wahlbeck, K. Glutamatergic drugs for schizophrenia: a systematic review and meta-analysis. Schizophr Res. 2005; 72(2–3): 225234.CrossRefGoogle ScholarPubMed
21. Ventura, J, Subotnik, KL, Gitlin, MJ, et al. Negative symptoms and functioning during the first year after a recent onset of schizophrenia and 8 years later. Schizophr Res. 2015; 161(2–3): 407413. Epub ahead of print Dec 8, 2014. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4308531/. Accessed January 13, 2017.CrossRefGoogle ScholarPubMed
22. Remberk, B, Bazynska, AK, Bronowska, Z, et al. Which aspects of long-term outcome are predicted by positive and negative symptoms in early-onset psychosis? An exploratory eight-year follow-up study. Psychopathology. 2015; 48(1): 4755. Epub ahead of print Dec 2, 2014.CrossRefGoogle ScholarPubMed
23. Marchesi, C, Affaticati, A, Monici, A, De Panfilis, C, Ossola, P, Tonna, M. Severity of core symptoms in first episode schizophrenia and long-term remission. Psychiatry Res. 2015; 225(1–2): 129132. Epub ahead of print Nov 11, 2014.CrossRefGoogle ScholarPubMed
24. Galderisi, S, Bucci, P, Mucci, A, et al. Categorical and dimensional approaches to negative symptoms of schizophrenia: focus on long-term stability and functional outcome. Schizophr Res. 2013; 147(1): 157162. Epub ahead of print Apr 19.Google Scholar
25. Agid, O, Siu, CO, Pappadopulos, E, Vanderburg, D, Remington, G.. Early prediction of clinical and functional outcome in schizophrenia. Eur Neuropsychopharmacol. 2013; 23(8): 842851. Epub ahead of print Nov 7, 2012.Google Scholar
26. Hofer, A, Baumgartner, S, Bodner, T, et al. Patient outcomes in schizophrenia, II: the impact of cognition. Eur Psychiatry. 2005; 20(5–6): 395402.CrossRefGoogle ScholarPubMed
27. Zhang, L, Zhao, J. Profile of minocycline and its potential in the treatment of schizophrenia. Neuropsychiatr Dis Treat. 2014; 10: 11031111. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4069141/. Accessed January 13, 2017.Google Scholar
28. Monji, A, Kato, TA, Mizoguchi, Y, et al. Neuroinflammation in schizophrenia especially focused on the role of microglia. Prog Neuropsychopharmacol Biol Psychiatry. 2013; 42: 115121. Epub ahead of print Dec 13, 2011.Google Scholar
29. Glantz, LA, Gilmore, JH, Lieberman, JA, Jarskog, LF. Apoptotic mechanisms and the synaptic pathology of schizophrenia. Schizophr Res. 2006; 81(1): 4763. Epub ahead of print Oct 14, 2005.CrossRefGoogle ScholarPubMed
30. Sertan Copoglu, U, Virit, O, Hanifi Kokacya, M, et al. Increased oxidative stress and oxidative DNA damage in non-remission schizophrenia patients. Psychiatry Res. 2015; 229(1–2): 200205. Epub ahead of print Jul 15.Google Scholar
31. Howes, O, McCutcheon, R, Stone, J. Glutamate and dopamine in schizophrenia: an update for the 21st century. J Psychopharmacol. 2015; 29(2): 97115. Epub ahead of print Jan 13. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4902122/. Accessed January 13, 2017.CrossRefGoogle Scholar
32. Xu, S, Gullapalli, RP, Frost, DO. Olanzapine antipsychotic treatment of adolescent rats causes long-term changes in glutamate and GABA levels in the nucleus accumbens. Schizophr Res. 2015; 161(2–3): 452457. Epub ahead of print Dec 5, 2014. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4308953/. Accessed January 13, 2017.CrossRefGoogle ScholarPubMed
33. Goldstein, ME, Anderson, VM, Pillai, A, Kydd, RR, Russell, BR. Glutamatergic neurometabolites in clozapine-responsive and -resistant schizophrenia. Int J Neuropsychopharmacol. 2015; 18(6): pii: pyu117. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4438552/. Accessed January 13, 2017.Google Scholar
34. Koprivica, V, Regardie, K, Wolff, C, et al. Aripiprazole protects cortical neurons from glutamate toxicity. Eur J Pharmacol. 2011; 651(1–3): 7376. Epub ahead of print Nov 18, 2010.Google Scholar
35. Millan, MJ, Fone, K, Steckler, T, Horan, WP. Negative symptoms of schizophrenia: clinical characteristics, pathophysiological substrates, experimental models and prospects for improved treatment. Eur Neuropsychopharmacol. 2014; 24(5): 645692. Epub ahead of print Apr 4. http://www.europeanneuropsychopharmacology.com/article/S0924-977X(14)00093-5/pdf. Accessed January 13, 2017.Google Scholar
36. Millan, MJ, Agid, Y, Brune, M, et al. Cognitive dysfunction in psychiatric disorders: characteristics, causes and the quest for improved therapy. Nat Rev Drug Discov. 2012; 11(2): 141168.CrossRefGoogle ScholarPubMed
37. Oya, K, Kishi, T, Iwata, N. Efficacy and tolerability of minocycline augmentation therapy in schizophrenia: a systematic review and meta-analysis of randomized controlled trials. Hum Psychopharmacol. 2014; 29(5): 483491. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5170618/. Accessed January 13, 2017.CrossRefGoogle ScholarPubMed
38. Iwata, Y, Nakajima, S, Suzuki, T, et al. Effects of glutamate positive modulators on cognitive deficits in schizophrenia: a systematic review and meta-analysis of double-blind randomized controlled trials. Mol Psychiatry. 2015; 20(10): 11511160. Epub ahead of print Jun 16.Google Scholar
39. Kay, SR, Fiszbein, A, Opler, LA. The positive and negative syndrome scale (PANSS) for schizophrenia. Schizophr Bull. 1987; 13(2): 261276. https://academic.oup.com/schizophreniabulletin. Accessed January 13, 2017.Google Scholar
40. Andreasen, NC. Negative symptoms in schizophrenia: definition and reliability. Arch Gen Psychiatry. 1982; 39(7): 784788.Google Scholar
41. Guy, W. ECDEU (Early Clinical Drug Evaluation) Assessment Manual for Psychopharmacology. Washington, DC: U.S. Department of Heath, Education, and Welfare Public Health Service Alcohol, Drug Abuse, and Mental Health Administration; 1976. https://archive.org/details/ecdeuassessmentm1933guyw. Accessed January 13, 2017.Google Scholar
42. Ghanizadeh, A, Dehbozorgi, S, OmraniSigaroodi, M, Rezaei, Z. Minocycline as add-on treatment decreases the negative symptoms of schizophrenia: a randomized placebo-controlled clinical trial. Recent Pat Inflamm Allergy Drug Discov. 2014; 8(3): 211215.Google Scholar
43. Kelly, DL, Sullivan, KM, McEvoy, JP, et al. Adjunctive Minocycline in Clozapine-Treated Schizophrenia Patients With Persistent Symptoms. J Clin Psychopharmacol. 2015; 35(4): 374381. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4485552/. Accessed January 13, 2017.CrossRefGoogle ScholarPubMed
44. Moher, D, Liberati, A, Tetzlaff, J, Altman, DG, The PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Int J Surg. 2010; 8(5): 336341. Epub ahead of print Feb 18. http://www.journal-surgery.net/article/S1743-9191(10)00040-3/pdf. Accessed January 13, 2017.Google Scholar
45. Overall, JE, Gorham, DR. The brief psychiatric rating scale. Psychol Rep. 1962; 10: 799812. http://www.statpower.net/Content/312/Homework/OverallGorham1962.pdf. Accessed January 13, 2017.CrossRefGoogle Scholar
46. Lancon, C, Auquier, P, Reine, G, Bernard, D, Toumi, M. Study of the concurrent validity of the Calgary Depression Scale for Schizophrenics (CDSS). J Affect Disord. 2000; 58(2): 107115.CrossRefGoogle ScholarPubMed
47. Hamilton, M. A rating scale for depression. J Neurol Neurosurg Psychiatry. 1960; 23: 5662. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC495331/. Accessed January 13, 2017.Google Scholar
48. Beck, AT, Alford, BA. Depression: Causes and Treatment, 2nd ed. Philadelphia: University of Pennsylvania Press; 2009.Google Scholar
49. Nuechterlein, KH, Green, MF, Kern, RS, et al. The MATRICS Consensus Cognitive Battery, part 1: test selection, reliability, and validity. Am J Psychiatry. 2008; 165(2): 203213. Epub ahead of print Jan 2. http://ajp.psychiatryonline.org/doi/pdf/10.1176/appi.ajp.2007.07010042. Accessed January 13, 2017.CrossRefGoogle ScholarPubMed
50. Green, MF, Nuechterlein, KH. The MATRICS initiative: developing a consensus cognitive battery for clinical trials. Schizophr Res. 2004; 72(1): 13.Google Scholar
51. CANTAB. Cambridge: Cambridge Cognition Ltd. http://www.cambridgecognition.com/. Accessed January 13, 2017.Google Scholar
52. Chouinard, G, Margolese, HC. Manual for the Extrapyramidal Symptom Rating Scale (ESRS). Schizophr Res. 2005; 76(2–3): 247265. Epub ahead of print Apr 18.Google Scholar
53. Higgins, JP, Altman, DG, Gotzsche, PC, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ. 2011; 343: d5928. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3196245/. Accessed January 13, 2017.Google Scholar
54. RevMan 5. http://tech.cochrane.org/revman. Accessed January 13, 2017.Google Scholar
55. DerSimonian, R, Kacker, R. Random-effects model for meta-analysis of clinical trials: an update. Contemp Clin Trials. 2007; 28(2): 105114. Epub ahead of print May 12, 2006.Google Scholar
56. DerSimonian, R, Laird, N. Meta-analysis in clinical trials. Control Clin Trials. 1986; 7(3): 177188.Google Scholar
57. Higgins, JP, Thompson, SG, Deeks, JJ, Altman, DG. Measuring inconsistency in meta-analyses. BMJ. 2003; 327(7414): 557560. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC192859/. Accessed January 13, 2017.CrossRefGoogle ScholarPubMed
58. Comprehensive Meta-Analysis (CMA). https://www.meta-analysis.com/?gclid=CO2P5tX3isoCFRThGwodz-8L9A. Accessed January 13, 2017.Google Scholar
59. Egger, M, Davey Smith, G, Schneider, M, Minder, C. Bias in meta-analysis detected by a simple, graphical test. BMJ. 1997; 315(7109): 629634. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2127453/. Accessed January 13, 2017.Google Scholar
60. Begg, CB, Mazumdar, M. Operating characteristics of a rank correlation test for publication bias. Biometrics. 1994; 50(4): 10881101.Google Scholar
61. Duval, S, Tweedie, R. Trim and fill: a simple funnel-plot-based method of testing and adjusting for publication bias in meta-analysis. Biometrics. 2000; 56(2): 455463.Google Scholar
62. Chaves, C, Marque, CR, Maia-de-Oliveira, JP, et al. Effects of minocycline add-on treatment on brain morphometry and cerebral perfusion in recent-onset schizophrenia. Schizophr Res. 2015; 161(2–3): 439445. Epub ahead of print Dec 12, 2014.Google Scholar
63. Lisiecka, DM, Suckling, J, Barnes, TR, et al. The benefit of minocycline on negative symptoms in early-phase psychosis in addition to standard care—extent and mechanism (BeneMin): study protocol for a randomised controlled trial. Trials. 2015; 16: 71. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4351843/. Accessed January 13, 2017.Google Scholar
64. Fekadu, A, Mesfin, M, Medhin, G, et al. Adjuvant therapy with minocycline for schizophrenia (the MINOS Trial): study protocol for a double-blind randomized placebo-controlled trial. Trials. 2013; 14: 406. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4222697/. Accessed January 13, 2017.CrossRefGoogle ScholarPubMed
65. Chaudhry, IB, Hallak, J, Husain, N, et al. Minocycline benefits negative symptoms in early schizophrenia: a randomised double-blind placebo-controlled clinical trial in patients on standard treatment. J Psychopharmacol. 2012; 26(9): 11851193. Epub ahead of print Apr 23. http://journals.sagepub.com/doi/pdf/10.1177/0269881112444941. Accessed January 13, 2017.Google Scholar
66. Khodaie-Ardakani, MR, Mirshafiee, O, Farokhnia, M, et al. Minocycline add-on to risperidone for treatment of negative symptoms in patients with stable schizophrenia: randomized double-blind placebo-controlled study. Psychiatry Res. 2014; 215(3): 540546. Epub ahead of print Jan 9.Google Scholar
67. Liu, F, Guo, X, Wu, R, et al. Minocycline supplementation for treatment of negative symptoms in early-phase schizophrenia: a double blind, randomized, controlled trial. Schizophr Res. 2014; 153(1–3): 169176. Epub ahead of print Feb 3.Google Scholar
68. Levkovitz, Y, Mendlovich, S, Riwkes, S, et al. A double-blind, randomized study of minocycline for the treatment of negative and cognitive symptoms in early-phase schizophrenia. J Clin Psychiatry. 2010; 71(2): 138149. Epub ahead of print Nov 3, 2009.CrossRefGoogle ScholarPubMed
69. Miyaoka, T, Yasukawa, R, Yasuda, H, Hayashida, M, Inagaki, T, Horiguchi, J. Minocycline as adjunctive therapy for schizophrenia: an open-label study. Clin Neuropharmacol. 2008; 31(5): 287292.Google Scholar
70. Kelly, DL, Vyas, G, Richardson, CM, et al. Adjunct minocycline to clozapine treated patients with persistent schizophrenia symptoms. Schizophr Res. 2011; 133(1–3): 257258. Epub ahead of print Aug 26.Google Scholar
71. Jhamnani, K, Shivakumar, V, Kalmady, S, Rao, NP, Venkatasubramanian, G. Successful use of add-on minocycline for treatment of persistent negative symptoms in schizophrenia. J Neuropsychiatry Clin Neurosci. 2013; 25(1): E06E07. http://neuro.psychiatryonline.org/doi/pdf/10.1176/appi.neuropsych.11120376. Accessed January 13, 2017.Google Scholar
72. Qurashi, I, Collins, J, Chaudhry, I, Husain, N. Promising use of minocycline augmentation with clozapine in treatment-resistant schizophrenia. J Psychopharmacol. 2014; 28(7): 707708. Epub ahead of print Mar 19. http://journals.sagepub.com/doi/pdf/10.1177/0269881114527358. Accessed January 13, 2017.Google Scholar
73. Carbon, M, Correll, CU. Thinking and acting beyond the positive: the role of the cognitive and negative symptoms in schizophrenia. CNS Spectr. 2014; 19(Suppl. 1): 3852; quiz 35–37, 53.Google Scholar
74. Carvalho, AF, Miskowiak, KK, Hyphantis, TN, et al. Cognitive dysfunction in depression: pathophysiology and novel targets. CNS Neurol Disord Drug Targets. 2014; 13(10): 18191835.Google Scholar
75. Kane, JM, Kishimoto, T, Correll, CU. Non-adherence to medication in patients with psychotic disorders: epidemiology, contributing factors and management strategies. World Psychiatry. 2013; 12(3): 216226. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3799245/. Accessed January 13, 2017.Google Scholar
76. Tehrani, R, Nash-Goelitz, A, Adams, E, Dahiya, M, Eilers, D. Minocycline-induced cutaneous polyarteritis nodosa. J Clin Rheumatol. 2007; 13(3): 146149.Google Scholar
77. Ramakrishna, J, Johnson, AR, Banner, BF. Long-term minocycline use for acne in healthy adolescents can cause severe autoimmune hepatitis. J Clin Gastroenterol. 2009; 43(8): 787790.Google Scholar
78. Benjamin, RW, Calikoglu, AS. Hyperthyroidism and lupus-like syndrome in an adolescent treated with minocycline for acne vulgaris. Pediatr Dermatol. 2007; 24(3): 246249.Google Scholar
79. Ahmed, F, Kelsey, PR, Shariff, N. Lupus syndrome with neutropenia following minocycline therapy: a case report. Int J Lab Hematol. 2008; 30(6): 543545.Google Scholar
Figure 0

Figure 1 PRISMA flowchart.

Figure 1

Table 1 Study, patient, illness and treatment characteristics on the meta-analyzed studies

Figure 2

Table 2 Meta-analysis and publication bias of efficacy and cognitive outcomes

Figure 3

Table 3 Meta-regression analysis of the heterogeneous findings

Supplementary material: File

Solmi supplementary material

Table S1

Download Solmi supplementary material(File)
File 35.8 KB
Supplementary material: File

Solmi supplementary material

Table S2

Download Solmi supplementary material(File)
File 15.2 KB