Next Article in Journal
High Myopia and Thickness of Extraocular and Masticatory Muscles—7T MRI, Preliminary Study
Next Article in Special Issue
Unruptured Anterior Communicating Artery Aneurysms: Management Strategy and Results of a Single-Center Experience
Previous Article in Journal
Pneumatic Compression Combined with Standard Treatment after Total Hip Arthroplasty and Its Effects on Edema of the Operated Limb and on Physical Outcomes: A Pilot Clinical Randomized Controlled Study
Previous Article in Special Issue
Distal Flow Diversion with Anti-Thrombotically Coated and Bare Metal Low-Profile Flow Diverters—A Comparison
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Inflammatory Drug Therapy in Aneurysmal Subarachnoid Hemorrhage: A Systematic Review and Meta-Analysis of Prospective Randomized and Placebo-Controlled Trials

Department of Neurosurgery, University Hospital Leipzig, 04103 Leipzig, Germany
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(12), 4165; https://doi.org/10.3390/jcm12124165
Submission received: 26 April 2023 / Revised: 2 June 2023 / Accepted: 17 June 2023 / Published: 20 June 2023
(This article belongs to the Special Issue Clinical Advances in Cerebral Aneurysm Treatment)

Abstract

:
Emerging evidence suggests that neuroinflammation may play a potential role in aneurysmal subarachnoid hemorrhage (aSAH). We aim to analyze the influence of anti-inflammatory therapy on survival and outcome in aSAH. Eligible randomized placebo-controlled prospective trials (RCTs) were searched in PubMed until March 2023. After screening the available studies for inclusion and exclusion criteria, we strictly extracted the main outcome measures. Dichotomous data were determined and extracted by odds ratio (OR) with 95% confidence intervals (CIs). Neurological outcome was graded using the modified Rankin Scale (mRS). We created funnel plots to analyze publication bias. From 967 articles identified during the initial screening, we included 14 RCTs in our meta-analysis. Our results illustrate that anti-inflammatory therapy yields an equivalent probability of survival compared to placebo or conventional management (OR: 0.81, 95% CI: 0.55–1.19, p = 0.28). Generally, anti-inflammatory therapy trended to be associated with a better neurologic outcome (mRS ≤ 2) compared to placebo or conventional treatment (OR: 1.48, 95% CI: 0.95–2.32, p = 0.08). Our meta-analysis showed no increased mortality form anti-inflammatory therapy. Anti-inflammatory therapy in aSAH patients tends to improve the neurological outcome. However, multicenter, rigorous, designed, prospective randomized studies are still needed to investigate the effect of fighting inflammation in improving neurological functioning after aSAH.

1. Introduction

Aneurysmal subarachnoid hemorrhage (aSAH) is a severe neurological disorder that affects 6–9 individuals per 100,000 each year and has a mortality rate of 35% [1]. In recent decades, the incidences of aSAH have decreased globally due to reduced smoking rates and improved management of arterial hypertension [2]. However, mortality and morbidity rates remain high, and survivors often experience poor functional outcomes, with roughly half unable to return to their previous life [3]. Prehospital mortality rates from aSAH are still between 22% and 26% [4]. Additionally, about 35% of aSAH patients suffer from memory disturbances, depression, and reduced quality of life [5,6,7,8].
Cerebral vasospasm (CVS) is linked to delayed cerebral ischemia (DCI) and is believed to play a role in unfavorable results following aSAH [9]. In order to enhance outcomes, various rescue therapies are utilized, including the selective intra-arterial infusion of vasodilators, balloon angioplasty, and induced hypertension [3,10]. However, the underlying pathophysiological mechanisms that enable injury expansion following aSAH are still not fully understood, and only a limited number of pharmacological treatments have been proven effective. Recent data suggest that neuroinflammation has a pivotal effect in injury expansion and neurological deficits [11,12,13]. Immune cells from the periphery are recruited into the brain parenchyma, and their activated forms release inflammatory cytokines [14,15]. Vessels affected by cerebral vasospasm have been found to have an elevated leukocyte adhesion capacity, resulting in delayed neurologic deterioration [16,17].
Numerous previous clinical trials have investigated the use of various anti-inflammatory drugs in aSAH patients to improve mortality and functional outcomes. In this meta-analysis, we aim to examine the available evidence and identify effective drug interventions that could improve functional outcomes and survival rates in aSAH patients.

2. Materials and Methods

To carry out this systematic review, we utilized the Cochrane Collaboration format [18] and adhered to the PRISMA checklist (see Supplementary Figure S1) [19]. The study was registered in the “International Prospective Register of Systematic Reviews” (PROSPERO) in 2023 (CRD42023395375), and a comprehensive, predetermined protocol can be obtained upon request.

2.1. Search Strategy for Identification of Studies

The authors performed a systematic search of the Pubmed database (http://www.ncbi.nlm.nih.gov/pubmed) using the terms “aneurysmal subarachnoid hemorrhage”, “SAH”, and “subarachnoid hemorrhage”. The search was limited to randomized controlled trials, human studies, and English-language articles. The authors screened the articles for randomized placebo, controlled trials investigating anti-inflammatory drug therapies. The literature search encompassed all results until 31 March 2023. The inclusion criteria were developed based on the PICOS (population, intervention, comparator, outcomes, and study design) framework [20]. The following criteria were applied: patients suffered from aSAH; anti-inflammatory drug therapies were implemented; results were compared to a placebo arm; all results of the prespecified endpoints were reported; and the trials were defined as prospective, randomized, placebo-controlled, and double-blinded. The authors excluded certain types of results, such as reviews, letters, study protocols, conference abstracts, unpublished manuscripts, animal experiments, and studies with insufficient data (e.g., randomized controlled trials without a placebo arm). Furthermore, we conducted a search for studies that matched our inclusion and exclusion criteria in previous meta-analyses and systematic reviews. The identified articles underwent a stepwise evaluation process: Initial screening of study titles, followed by assessment of corresponding abstracts, and in case of uncertainty, the full-text screening by two authors (JW and EG) until all retrieved studies were either included or excluded.

2.2. Types of Studies

Randomized, double-blind, placebo-controlled clinical trials that evaluated the efficacy of anti-inflammatory treatment in improving the outcome and survival rates of aneurysmal subarachnoid hemorrhage were included. Trials without survival or neurological outcome data were excluded. Trials that specifically investigated elderly or geriatric aSAH patients (>60 years) were excluded. Neurological outcome was measured using the modified Rankin Scale (mRS). If an article presented numerical data for individual mRS classes, the data were documented based on the definition of a poor outcome as a score ranging from 3 to 6 on the mRS scale, while a score ≤ 2 was considered indicative of a good outcome [21]. If a study presented the percentage of patients with a defined outcome event, the absolute numbers were computed using the provided percentages.

2.3. Data Extraction and Quality Evaluation

Baseline data extracted from the studies included the study names, first authors, publication year, country, number of centers (whether mono-, bi-, or multicentric), key trial design characteristics (such as randomization method, allocation method, presence of missing data, lack of reported outcome, and utilization of intention-to-treat analysis), as well as other pertinent information.

2.4. Statistics

The meta-analyses were performed using Review Manger Web (RevMan Web Version 5.4.1 from The Cochrane Collaboration). Statistical heterogeneity was assessed using the x2, while inconsistency was evaluated using the I2 statistics. Substantial heterogeneity was considered when the I2 value reached 50% or higher. The weight of each individual trial’s contribution to the treatment effect estimation was determined based on the study sample size. Data from a multi-arm trial that involved different dosage regimens were combined to create a unified group for inclusion in the pairwise meta-analysis [22]. To assess publication bias for the defined endpoints of survival and neurological outcome, three methods were applied. Firstly, funnel plots were generated to visually examine the publication bias of the included trials. Secondly, an Egger regression analysis was conducted to statistically evaluate the symmetry of the funnel plot. The likelihood of publication bias was determined using the two-tailed Egger regression intercept test, with a significance threshold set at 5% [23]. Thirdly, Begg’s test was utilized to evaluate the symmetry of the data [24]. MedCalc (Version 20.123 for Windows) was used to perform both Egger’s and Begg’s tests. Pooled odds ratio (OR) estimates, using a random effects model, were used to express the effect sizes. Analyses were conducted for both death and poor outcome.

3. Results

3.1. Literature Search

Based on the search strategy, a total of 967 English articles (Figure 1) were deemed eligible. Following the evaluation of the titles, abstracts, and full texts, 953 articles were excluded. Ultimately, 14 articles encompassing 1759 patients were included for the meta-analysis.

3.2. Characteristics of Included Studies

The included studies were published from 1989 to 2022. Table 1 shows the major characteristics of the anti-inflammatory drug interventions of the 14 included trials [25,26,27,28,29,30,31,32,33,34,35,36,37,38]. The participants included in each study were all aneurysmal SAH patients. The duration of treatments varied from 24 h to 21 days. One prospective, double-blind, and placebo-controlled trial was designed as a multi-arm trial [34]. Data were included by combining groups into the pairwise meta-analysis of our dichotomous endpoints. Further identified anti-inflammatory prospective studies in aSAH that did not fulfill our inclusion criteria due to single-blinded design, unknown blinding method, investigating elderly patients only, written in other languages than English, unavailable data for the primary endpoints of this meta-analysis, or the lack of placebo arms are provided in the Supplementary Table S1 [39,40,41,42,43,44,45,46,47,48,49,50,51].

3.3. Risk of Bias and Quality Assessment

All included trials used a double-blinded design to minimize performance bias and detection bias of the personnel and patients. Furthermore, all trials were defined as randomized trials. The randomization technique was not reported in seven trials [25,26,27,29,30,31,34]. All prespecified endpoints are reported with the corresponding outcomes. Generally, no prospective, randomized, double-blind, and placebo-controlled trial showed characteristics that might indicate a high risk of bias. The frequency of the individual bias of each trial and the overall bias assessment are displayed in Figure 2. The detailed quality assessment protocol regarding risk of bias and author judgments for the individual studies are given in Supplementary Table S2.

3.4. Impact of Anti-Inflammatory Therapy in Aneurysmal SAH on Survival

A total of 13 studies met our selection criteria and reported data regarding survival [25,26,27,28,29,30,32,33,34,35,36,37,38]. A total of 1689 patients were randomized into either anti-inflammatory (n = 885) or placebo treatment (n = 804). A total of 78 patients (8.8%) out of 885 in the anti-inflammatory treatment arm died, and 85 (10.6%) of the 804 patients in the control arms died. The outcome evaluation ranged from one month to one year after treatment. The overall odds ratio (Figure 3) for death in the pooled analysis was 0.81 (95% CI: 0.55–1.19) (p = 0.28). No significant heterogeneity was present (I2 = 8%, p = 0.36). No studies were identified that indicated a significant positive or negative impact of anti-inflammatory therapy on the outcome endpoint of “death”.

3.5. Impact of Anti-Inflammatory Therapy in Aneurysmal SAH on Neurological Outcome

A total of 9 studies met our selection criteria and reported data regarding neurological outcome, which enabled a dichotomization into mRS ≤ 2 or mRS > 2 [25,26,27,29,30,31,32,35,38]. A total of 1223 patients were randomized into either anti-inflammatory (n = 601) or control treatment (n = 622). In total, 428 patients (71.2%) out of 601 in the anti-inflammatory treatment arm achieved a good neurological outcome (mRS ≤ 2), and 419 (67.4%) of the 622 patients in the placebo arms achieved a favorable outcome. The outcome evaluation ranged from one month to one year after treatment. The overall odds ratio (Figure 4) for mRS ≤ 2 in the pooled analysis was 1.48 (95% CI: 0.95–2.32) (p = 0.08). No significant heterogeneity was present (I2 = 45%, p = 0.07). One study investigating the effect of dapsone reported a beneficial effect regarding functional outcome as endpoint [27]. Furthermore, cerebrolysin also resulted in an increased probability to achieve a favorable outcome compared to placebo treatment [38].

3.6. Publication Bias

In order to ensure a reliable assessment, several measures were implemented to examine the presence of publication bias. Firstly, a comprehensive literature search strategy was applied. Secondly, the selected studies included in this meta-analysis strictly adhered to the predefined inclusion and exclusion criteria. Thirdly, publication bias was evaluated using funnel plots (see Figure 5) for the endpoints (survival and favorable mRS outcome (≤2)). Finally, statistical analyses of publication bias using Egger´s and Begg´s tests were performed. The data points for survival analysis were all positioned within the inverted funnel, indicating no visually apparent significant publication bias in relation to the survival analysis.
Following the visual examination for publication bias, both Egger’s and Begg’s tests were conducted to investigate the presence of publication bias for the primary outcomes. Regarding mortality, Egger´s test indicated a statistically significant publication bias (p = 0.03, intercept = −1.05, 95% CI −1.99–−0.11), while Begg´s test revealed a Kendall’s tau of −0.23 (p = 0.27). We identified one study investigating dapsone in aSAH with a very high effect size in the bottom-right corner of the funnel plot regarding favorable mRS outcome analysis [26]. Hence, there might be the risk that there is an underbelly of unpublished studies with similar standard errors, but small and non-significant effects. Furthermore, Egger´s test did not indicate any significant publication bias concerning the endpoint of “favourable mRS outcome (≤2)” (p = 0.10, intercept = 1.23, 95% CI −0.30–2.76), and Begg’s test yielded a Kendall’s tau of 0.29 (p = 0.25).

4. Discussion

The present meta-analysis summarized available data on anti-inflammatory therapy in aSAH and suggests a potential need for further trials investigating anti-inflammatory therapies using dapsone or corticosteroids. These drugs may positively influence the neurological outcome after aSAH. Our results indicate that anti-inflammatory therapy may have a positive effect on functional outcome. This provides valuable evidence for clinicians when considering the prescription of effective medications for patients with aSAH. Nimodipine remains the benchmark medical drug therapy for preventing DCI and influencing the outcome in aSAH patients [3,52]. In the following section, we will discuss the findings of the present meta-analysis and debate the classes of investigated anti-inflammatory drugs.

4.1. COX Inhibition

Cyclooxygenase (COX) is a bifunctional enzyme that acts as a cyclo-oxygenase and catalase. The conversion of arachidonic acid to prostaglandins is facilitated by the enzymes COX-1 and COX-2. COX-1 synthesizes thromboxane A2, which induces platelet aggregation, vasoconstriction, and proliferation of smooth muscles. In contrast, COX-2 synthesizes prostacyclin, which induces vasodilation and relaxation of smooth muscles and antagonizes thromboxane A2 in the macrovascular endothelium [53,54]. Animal investigations have shown that arterial endothelial cells have an increased expression of COX-2 after SAH, while the expression of COX-1 remains unchanged [55]. Animal trials have shown that inhibiting COX-2 may prevent brain edema and preserve neurological functioning [56]. However, non-selective COX-2 inhibition by nonsteroidal anti-inflammatory medications, such as acetylsalicylic acid, might elevate the risk of re-bleeding, despite attenuating neuroinflammation in aSAH [57,58]. Although acetylsalicylic acid failed to significantly reduce the risk of a delayed ischemic neurological deficit and improve the functional outcome in prospective, double-blind, and placebo-controlled trials [30,36], there was a tendency to prescribe it, as its relative risk reduction for poor outcome was 21% [36]. Celecoxib, a selective COX-2 inhibitor, has not been prospectively investigated in randomized and double-blinded aSAH trials yet. A small clinical trial on subjects with intracerebral hemorrhage showed that celecoxib reduced hematoma and edema volume through its anti-inflammatory effect [59]. However, the current limited evidence for celecoxib therapy in aSAH is due to the failure of acetylsalicylic acid and the known increased risk of myocardial infarction with celecoxib therapy [60].

4.2. Thromboxane A2 Synthetase Inhibition

Thromboxane A2 (TXA2) is a prothrombotic prostanoid that is predominantly synthesized via COX-1, COX-2, and TXA2 synthase [61]. Furthermore, it is released by activated platelets in the case of inflammation or tissue injury [62]. Afterwards, TXA2 binds to the TXA2 receptor, resulting in smooth muscle cell constriction and platelet aggregation. Experimental SAH rat models showed that the SAH-induced regional blood flow reduction is closely linked to an increased expression of TXA2 receptors in the smooth muscle cells of cerebral arteries and microvessels [63]. An increase of TXA2 levels induces leukocyte aggregation and systemic inflammation. However, the already debated acetylsalicylic acid can reduce the TXA2 levels by inactivating COX-1 [64], whereas nimodipine exerts no effect on serum thromboxane levels [65]. The prospective randomized double-blind trial of Suzuki et al. [34] showed a potential use of a thromboxane synthetase inhibitor in preventing vasospasm and infarcts in severe aSAH cases with Hunt & Hess grades III and IV. However, the results of this trial regarding functional outcome could not be included in the statistical analysis because they were not transferrable to the mRS scale.

4.3. Epoxide Hydrolase Inhibition

Soluble epoxide hydrolase is the metabolizing enzyme of epoxyeicosatrienoic acids, which oppose the expression of vascular cell adhesion molecule-1 (VCAM-1) by blocking the NF-κB translocation. The translocation NF-κB is an important part of the expression of the pro-inflammatory adhesion molecule VCAM-1 [66,67]. The consequence of this vascular inflammatory pathway is the development of a vasogenic edema, causing the extravasation of ions and proteins through a disturbed blood–brain barrier [68]. Animal experiments have shown that the genetic deletion or inhibition of soluble epoxide hydrolase results in a reduced expression of VCAM-1 in the cerebrovasculature, thereby attenuating the transmigration of infiltrating inflammatory cells [69,70]. Increased levels of VCAM-1 have been observed in the plasma or CSF of aSAH patients, and this inflammatory response is suggested to be an important part of vasogenic edema development [69,71]. The first human prospective, randomized, and placebo-controlled trial investigating soluble epoxide hydrolase inhibition was recently conducted by Martini et al. [33], which showed that the application is safe for humans, and there was no drug-associated mortality. Furthermore, it was revealed that soluble epoxide hydrolase inhibition resulted in increased serum levels of epoxyeicosatrienoic acids, whereas the cerebrospinal fluid levels of epoxyeicosatrienoic acids were not sufficiently increased by the application of the soluble epoxide hydrolase inhibitor. To date, the current evidence is limited by the low power size, and the results of the present meta-analysis cannot recommend a routine clinical application. Further larger trials of soluble epoxide hydrolase inhibitors with an improved CNS penetration might also provide information regarding the functional outcome.

4.4. Statins

Statins, which are commonly referred to as HMG-CoA reductase inhibitors, have pleiotropic effects, including anti-inflammatory actions and the attenuation of vasospasm. Statins enhance the expression and activity of endothelial nitric oxide synthase and reduce nitric oxide scavenging by free radicals [72,73]. Moreover, they can inhibit the activation and migration of inflammatory perivascular granulocytes [74,75], potentially attenuating or preventing cerebral vasospasms after aSAH. To date, eight randomized [25,26,28,35,37,44,45,46] and four large multicenter, prospective, randomized trials [32,43,49,51] have evaluated the use of simvastatin, atorvastatin, pitavastatin, or pravastatin in aSAH patients. However, 2 studies had a special focus on either elderly patients over 60 years [39] or a dose comparison of simvastatin without a placebo group [51]. The primary endpoints of these studies were the frequency of vasospasm, presence of DCI, and the functional outcome. A previous meta-analysis, which summarized the results of some of the aforementioned trials focused on the potential reduction of vasospasm (defined by transcranial doppler sonography), found no significant effect [76]. However, another meta-analysis, which exclusively focused on statins and the treatment duration, found that a statin therapy for 2 weeks may improve neurological outcome. Surprisingly, prolonged use for 3 weeks had no significant effect on neurological outcome [77]. In the present meta-analysis, we also could not observe a significant effect on mortality or functional outcome. Current evidence and guidelines do not recommend routine use of statins in aSAH [3,77,78]. To assess the impact of statin treatment over a duration of two weeks, it is necessary to conduct future large-scale trials. The recently published AHA/ASA 2023 aSAH guideline does not recommend the routine use of statin therapy regarding the prevention of delayed cerebral ischemia [4].

4.5. Cerebrolysin

Cerebrolysin is a low-molecular-weight neuropeptide compound with free amino acids that is derived from the porcine brain tissue. Animal experiments have shown that cerbrolysin has anti-inflammatory properties that can be beneficial in cerebral ischemic stroke. By inducing the expression of anti-inflammatory factors and promoting microglia polarization towards an anti-inflammatory type, cerebrolysin was found to reduce the ischemic infarct volume [79]. Randomized placebo-controlled clinical studies investigating the effect of cerebrolysin in acute ischemic stroke have shown improved 3-month functional outcomes regarding the mRS [80,81,82,83,84,85]. A 10-year retrospective investigation of cerebrolysin treatment in severe aSAH patients found an increase in the survival rates at the 3-month mark for individuals who underwent surgical intervention for aSAH [86]. Woo et al. [38] provided the first results on cerebrolysin in aSAH patients from a prospective randomized double-blind trial. Cerebrolysin appeared to be a safe drug, and mortality was reduced in the cerebrolysin group compared to the placebo group. However, the study size did not allow for this conclusion, and the results regarding functional outcome were also neutral. However, future trials will need to explore cerebrolysin in a larger cohort and with an earlier intervention time-window because significant improvements in 3-month outcomes were found in trials investigating cerebrolysin in ischemic stroke when it was administered within 6 h after hospital admission [82,86].

4.6. Dapsone

Dapsone, also known as 4,4′-diamino-diphenylsulfone, is a neuroprotective compound that acts by suppressing glutamate-mediated excitotoxicity and possesses anti-inflammatory properties [87,88]. The anti-inflammatory properties of dapsone are attributed to its ability to hinder the calcium-dependent actions of neutrophils, which includes the suppression of harmful oxidants and proteases released by these cells, leading to tissue damage. Furthermore, dapsone irreversibly inhibits myeloperoxidase, blocks peroxidase functioning in eosinophils, inhibits the synthesis of products by 5-lipoxygenase products, and decreases the levels of neurotoxic free radicals [87,88,89,90]. A prospective randomized and double-blind trial investigating dapsone in acute ischemic stroke revealed that patients receiving dapsone had significant improved neurological functioning at 2 months after acute ischemic stroke compared to the placebo group [90]. To date, only one trial has analyzed the use of dapsone in aSAH patients [27]. Our meta-analysis revealed that this study showed the most promising effect in terms of the probability of a favorable outcome, as defined by the mRS. This beneficial effect appears to be mainly due to the protective role of dapsone in reducing the incidence of vasospasms and DCI. In the placebo group, DCI was present in 63.6% of the patients, while only 26.9% of the patients in the dapsone group had DCI [27]. Future multicenter trials will be necessary to confirm these findings and could also focus on physiological investigations of cerebral blood flow.

4.7. Corticosteroids

Corticosteroids are frequently used anti-inflammatory drugs in neurosurgical pathologies due to their diverse functioning as mineralocorticoid or anti-inflammatory drugs. However, there is currently no evidence or guideline supporting the use of corticosteroids to improve neurological outcomes after aSAH [91]. Guidelines suggest maintaining euvolemia in patients with symptomatic delayed cerebral ischemia to reduce the progression and severity of delayed cerebral ischemia, supported by class I evidence [3,4,92,93,94]. Early administration of fludrocortisone was shown to decrease natriuretic diuresis, maintain euvolemia, and reduce symptomatic vasospasm in a study by Nakagawa et al. [95]. Vasospasm is associated with the inflammatory response after aSAH [96], as shown by prospectively recorded serum interleukin-6 levels in 80 patients that were associated with higher Hunt & Hess grades, development of seizure, cerebral vasospasms, and chronic hydrocephalus [97]. Therefore, corticosteroid therapy is assumed to decrease vasospasm by attenuating the inflammatory response. Four investigations analyzed the impact of corticosteroids with minimal mineralocorticoid functions: two trials analyzed methylprednisolone [29,98], whereas two trials utilized dexamethasone [99,100]. Although one randomized controlled trial by Gomis et al. [29] showed a significant reduction of symptomatic vasospasm in high-grade aSAH patients, this effect did not result in a significant improvement of functional outcomes compared to the placebo group. However, the lower boundary of the 95% CI was 0.98 in our pooled analysis, suggesting at least a tendency towards the administration of corticosteroids. Schürkämper et al. [100] investigated dexamethasone in a retrospective study of 242 aSAH patients and found that high-dose (>12 mg/d or >5 days) dexamethasone treatment resulted in an increased probability of favorable outcome compared to those receiving a lower total dose (<12 mg/d or <5 days). However, these results are limited by the retrospective design, and the control group received low amounts of dexamethasone. The ongoing FINISHER trial (Fight INflammation to Improve Outcome after Aneurysmal Subarachnoid HEmorRhage, Trial-Number: NCT05132920) will provide the first data from a prospective randomized placebo-controlled trial evaluating dexamethasone. The primary endpoint of this multicentric trial is the mRS at 6 months after aSAH [101]. This study may elucidate whether the potential benefit of corticosteroids is based on the anti-inflammatory functioning. Furthermore, our current knowledge is based on a combination of several inflammatory pathways that contribute to the inflammatory response after aSAH. Dexamethasone may be an option to simultaneously inhibit several pathways (see Supplementary Figure S2), as the inhibition of single pathways has not shown a significant effect thus far.
This meta-analysis suggests a potential benefit of dapsone or corticosteroids in the management of aSAH, but the conclusions are limited by the low quality of evidence. Furthermore, the present meta-analysis of anti-inflammatory drugs is also limited by the fact that those drugs have different targets and effects. Hence, it is difficult to convex generalizability from the results of each individual study. Nevertheless, even the recent AHA/ASA 2023 aSAH guideline also points out that the emerging data suggesting inflammation to contribute to brain injury in aSAH necessitates further study on glucocorticoid steroids because their safety and efficacy profile is not sufficiently investigated so far [4]. Future studies will have to focus on whether there are different subgroups of aSAH patients who might benefit from different anti-inflammatory drugs targeting individual pathways. Additionally, the findings of the present meta-analysis investigating anti-inflammatory drugs must be interpreted with caution due to different pharmacodynamics and pharmacological effects. Several of the included anti-inflammatory drugs have also other functions, which are paramount regarding the care for aSAH patients. For instance, COX-1 inhibitors have also fundamental effects regarding the inhibition of platelet aggregation, whereas corticosteroids also act on the hemostatic system by increasing clotting factor levels and platelet counts [102,103,104]. The present analysis includes small studies with low statistical power and significant findings, while there are no studies with low power and non-significant results for mortality. Therefore, there may be a publication bias. A major limitation of the evaluation of the neurological outcome are the heterogeneous timepoints of the endpoints of the included studies and the different grades of clinical severity of aSAH, which confound our results. Ultimately, more rigorous data from large, prospective, randomized controlled trials are needed to evaluate the efficacy of dapsone or dexamethasone in the management of aSAH.

5. Conclusions

There was no difference in mortality between conventional therapy and anti-inflammatory therapy in patients with aneurysmal subarachnoid hemorrhage. However, certain individual anti-inflammatory therapies, such as dapsone and corticosteroids, may have a positive impact on functional outcomes. To establish sufficient evidence regarding the efficacy of anti-inflammatory therapy in aSAH, further multicenter, randomized, double-blind, and placebo-controlled trials are needed.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm12124165/s1, Table S1: Characteristics of excluded anti-inflammatory studies; Table S2: Quality assessment of prospective randomized double-blind, and placebo-controlled trials; Figure S1: PRISMA checklist summarizing the checklist items and the location of the items in the present meta-analysis [105], Figure S2: Illustrative summary of the potential anti-inflammatory functioning and avenues influenced by corticosteroid therapy in aneurysmal subarachnoid hemorrhage.

Author Contributions

Conceptualization, E.G. and J.W.; methodology, E.G. and J.W.; data curation, E.G. and J.W.; writing—original draft preparation, E.G. and J.W.; writing—review and editing, A.G., M.V., M.V. and E.G.; visualization, E.G. and J.W.; supervision, E.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study was conducted according to the guidelines of the Declaration of Helsinki and no approval was necessary in case of a meta-analysis.

Informed Consent Statement

Written informed consent was not needed due to the retrospective design. The local ethics committee waived patient informed consent for this retrospective observational study.

Data Availability Statement

All data are included in this manuscript.

Acknowledgments

The graphical abstract was created using BioRender. The authors thank Jenny Messall for providing assistance in the design of Supplementary Figure S2. The present publication was funded by the Open Access Publishing Fund of Leipzig University supported by the German Research Foundation within the program Open Access Publation Funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Neifert, S.N.; Chapman, E.K.; Martini, M.L.; Shuman, W.H.; Schupper, A.J.; Oermann, E.K.; Mocco, J.; Macdonald, R.L. Aneurysmal Subarachnoid Hemorrhage: The Last Decade. Transl. Stroke Res. 2020, 12, 428–446. [Google Scholar] [CrossRef]
  2. Etminan, N.; Chang, H.S.; Hackenberg, K.; de Rooij, N.K.; Vergouwen, M.D.I.; Rinkel, G.J.E.; Algra, A. Worldwide Incidence of Aneurysmal Subarachnoid Hemorrhage According to Region, Time Period, Blood Pressure, and Smoking Prevalence in the Population: A Systematic Review and Meta-analysis. JAMA Neurol. 2019, 76, 588–597. [Google Scholar] [CrossRef]
  3. Connolly, E.S., Jr.; Rabinstein, A.A.; Carhuapoma, J.R.; Derdeyn, C.P.; Dion, J.; Higashida, R.T.; Hoh, B.L.; Kirkness, C.J.; Naidech, A.M.; Ogilvy, C.S.; et al. Guidelines for the management of aneurysmal subarachnoid hemorrhage: A guideline for healthcare professionals from the American Heart Association/american Stroke Association. Stroke 2012, 43, 1711–1737. [Google Scholar] [CrossRef] [Green Version]
  4. Hoh, B.L.; Ko, N.U.; Amin-Hanjani, S.; Chou, S.H.-Y.; Cruz-Flores, S.; Dangayach, N.S.; Derdeyn, C.P.; Du, R.; Hänggi, D.; Hetts, S.W.; et al. 2023 Guideline for the Management of Patients with Aneurysmal Subarachnoid Hemorrhage: A Guideline From the American Heart Association/American Stroke Association. Stroke 2023. [Google Scholar] [CrossRef]
  5. Tjahjadi, M.; Heinen, C.; König, R.; Rickels, E.; Wirtz, C.R.; Woischneck, D.; Kapapa, T. Health-Related Quality of Life after Spontaneous Subarachnoid Hemorrhage Measured in a Recent Patient Population. World Neurosurg. 2013, 79, 296–307. [Google Scholar] [CrossRef] [PubMed]
  6. Taufique, Z.; May, T.; Meyers, E.; Falo, C.; Mayer, S.A.; Agarwal, S.; Park, S.; Connolly, E.S.; Claassen, J.; Schmidt, J.M. Predictors of Poor Quality of Life 1 Year after Subarachnoid Hemorrhage. Neurosurgery 2016, 78, 256–264. [Google Scholar] [CrossRef]
  7. Al-Khindi, T.; Macdonald, R.L.; Schweizer, T.A. Cognitive and Functional Outcome after Aneurysmal Subarachnoid Hemorrhage. Stroke 2010, 41, e519–e536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Eagles, M.E.; Tso, M.K.; Macdonald, R.L. Cognitive impairment, functional outcome, and delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. World Neurosurg. 2019, 124, e558–e562. [Google Scholar] [CrossRef]
  9. Roos, Y.B.; de Haan, R.J.; Beenen, L.F.; Groen, R.J.; Albrecht, K.W.; Vermeulen, M. Complications and outcome in patients with aneurysmal subarachnoid haemorrhage: A prospective hospital based cohort study in the Netherlands. J. Neurol. Neurosurg. Psychiatry 2000, 68, 337–341. [Google Scholar] [CrossRef] [Green Version]
  10. Platz, J.; Güresir, E.; Vatter, H.; Berkefeld, J.; Seifert, V.; Raabe, A.; Beck, J. Unsecured intracranial aneurysms and induced hypertension safe? Neurocrit. Care 2011, 14, 168–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Caffes, N.; Kurland, D.B.; Gerzanich, V.; Simard, J.M. Glibenclamide for the treatment of ischemic and hemorrhagic stroke. Int. J. Mol. Sci. 2015, 16, 4973–4984. [Google Scholar] [CrossRef] [Green Version]
  12. Makino, H.; Tada, Y.; Wada, K.; Liang, E.I.; Chang, M.; Mobashery, S.; Kanematsu, Y.; Kurihara, C.; Palova, E.; Kanematsu, M.; et al. Pharmacological stabilization of intracranial aneurysms in mice: A feasibility study. Stroke 2012, 43, 2450–2456. [Google Scholar] [CrossRef] [Green Version]
  13. Güresir, E.; Gräff, I.; Seidel, M.; Bauer, H.; Coch, C.; Diepenseifen, C.; Dohmen, C.; Engels, S.; Hadjiathanasiou, A.; Heister, U.; et al. Aneurysmal Subarachnoid Hemorrhage during the Shutdown for COVID-19. J. Clin. Med. 2022, 11, 2555. [Google Scholar] [CrossRef]
  14. Moraes, L.; Grille, S.; Morelli, P.; Mila, R.; Trias, N.; Brugnini, A.; Lluberas, N.; Biestro, A.; Lens, D. Immune cells subpopulations in cerebrospinal fluid and peripheral blood of patients with Aneurysmal Subarachnoid Hemorrhage. Springerplus 2015, 4, 195. [Google Scholar] [CrossRef] [Green Version]
  15. Kooijman, E.; Nijboer, C.H.; van Velthoven, C.T.; Kavelaars, A.; Kesecioglu, J.; Heijnen, C.J. The rodent endovascular puncture model of subarachnoid hemorrhage: Mechanisms of brain damage and therapeutic strategies. J. Neuroinflamm. 2014, 11, 2. [Google Scholar] [CrossRef] [Green Version]
  16. Provencio, J.J.; Badjatia, N. Participants in the International Multi-disciplinary Consensus Conference on Multimodality Monitoring. Monitoring inflammation (including fever) in acute brain injury. Neurocrit. Care 2014, 21 (Suppl. 2), S177–S186. [Google Scholar] [CrossRef]
  17. Xu, H.-L.; Garcia, M.; Testai, F.; Vetri, F.; Barabanova, A.; Pelligrino, D.A.; Paisansathan, C. Pharmacologic blockade of vascular adhesion protein-1 lessens neurologic dysfunction in rats subjected to subarachnoid hemorrhage. Brain Res. 2014, 1586, 83–89. [Google Scholar] [CrossRef] [PubMed]
  18. Higgins, J.; Green, S. Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0.; Cochrane Collab: London, UK, 2011; Available online: www.cochrane-handbook.org (accessed on 1 September 2022).
  19. Liberati, M.; Tetzlaff, J.; Altman, D.G.; PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 2009, 6, e1000097. [Google Scholar] [CrossRef] [PubMed]
  20. Schardt, C.; Adams, M.B.; Owens, T.; Keitz, S.; Fontelo, P. Utilization of the PICO framework to improve searching PubMed for clinical questions. BMC Med. Inform. Decis. Mak. 2007, 7, 16. [Google Scholar] [CrossRef] [Green Version]
  21. Lampmann, T.; Hadjiathanasiou, A.; Asoglu, H.; Wach, J.; Kern, T.; Vatter, H.; Güresir, E. Early Serum Creatinine Levels after Aneurysmal Subarachnoid Hemorrhage Predict Functional Neurological Outcome after 6 Months. J. Clin. Med. 2022, 11, 4753. [Google Scholar] [CrossRef] [PubMed]
  22. Rücker, G.; Cates, C.J.; Schwarzer, G. Methods for including information from multi-arm trials in pairwise meta-analysis. Res. Synth. Methods 2017, 8, 392–403. [Google Scholar] [CrossRef] [Green Version]
  23. Egger, M.; Smith, G.D.; Schneider, M.; Minder, C. Bias in meta-analysis detected by a simple, graphical test. BMJ 1997, 315, 629–634. [Google Scholar] [CrossRef] [Green Version]
  24. Begg, C.B.; Mazumdar, M. Operating Characteristics of a Rank Correlation Test for Publication Bias. Biometrics 1994, 50, 1088–1101. [Google Scholar] [CrossRef] [PubMed]
  25. Chou, S.H.; Smith, E.E.; Badjatia, N.; Nogueira, R.G.; Sims, J.R., 2nd; Ogilvy, C.S.; Rordorf, G.A.; Ayata, C. A randomized, double-blind, placebo-controlled pilot study of simvastatin in aneurysmal subarachnoid hemorrhage. Stroke 2008, 39, 2891–2893. [Google Scholar] [CrossRef] [Green Version]
  26. Diringer, M.N.; Dhar, R.; Scalfani, M.; Zazulia, A.R.; Chicoine, M.; Powers, W.J.; Derdeyn, C.P. Effect of High-Dose Simvastatin on Cerebral Blood Flow and Static Autoregulation in Subarachnoid Hemorrhage. Neurocrit. Care 2015, 25, 56–63. [Google Scholar] [CrossRef] [Green Version]
  27. García-Pastor, C.; de Llano, J.P.N.-G.; Balcázar-Padrón, J.C.; Tristán-López, L.; Rios, C.; Díaz-Ruíz, A.; Rodríguez-Hernandez, L.A.; Nathal, E. Neuroprotective effect of dapsone in patients with aneurysmal subarachnoid hemorrhage: A prospective, randomized, double-blind, placebo-controlled clinical trial. Neurosurg. Focus 2022, 52, E12. [Google Scholar] [CrossRef]
  28. Garg, K.; Sinha, S.; Kale, S.S.; Chandra, P.S.; Suri, A.; Singh, M.M.; Kumar, R.; Sharma, M.S.; Pandey, R.M.; Sharma, B.S.; et al. Role of simvastatin in prevention of vasospasm and improving functional outcome after aneurysmal subarachnoid hemorrhage: A prospective, randomized, double-blind, placebo-controlled pilot trial. Br. J. Neurosurg. 2013, 27, 181–186. [Google Scholar] [CrossRef]
  29. Gomis, P.; Graftieaux, J.P.; Sercombe, R.; Hettler, D.; Scherpereel, B.; Rousseaux, P. Randomized, double-blind, placebo-controlled, pilot trial of high-dose methylprednisolone in aneurysmal subarachnoid hemorrhage. J. Neurosurg. 2010, 112, 681–688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Hop, J.W.; Rinkel, G.J.E.; Algra, A.; van der Sprenkel, J.B.; van Gijn, J. Randomized pilot trial of postoperative aspirin in subarachnoid hemorrhage. Neurology 2000, 54, 872–878. [Google Scholar] [CrossRef] [PubMed]
  31. Katayama, Y.; Haraoka, J.; Hirabayashi, H.; Kawamata, T.; Kawamoto, K.; Kitahara, T.; Kojima, J.; Kuroiwa, T.; Mori, T.; Moro, N.; et al. A randomized controlled trial of hydrocortisone againast hyponatremia in patients with aneurysmal subarachnoid hemorrhage. Stroke 2007, 38, 2373–2375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kirkpatrick, P.J.; Turner, C.L.; Smith, C.; Hutchinson, P.J.; Murray, G.D.; STASH Collaborators. Simvastatin in aneurysmal subarachnoid haemorrhage (STASH): A multicentre randomised phase 3 trial. Lancet Neurol. 2014, 13, 666–675. [Google Scholar] [CrossRef]
  33. Martini, R.P.; Siler, D.; Cetas, J.; Alkayed, N.J.; Allen, E.; Treggiari, M.M. A Double-Blind, Randomized, Placebo-Controlled Trial of Soluble Epoxide Hydrolase Inhibition in Patients with Aneurysmal Subarachnoid Hemorrhage. Neurocrit. Care 2022, 36, 905–915. [Google Scholar] [CrossRef]
  34. Suzuki, S.; Sano, K.; Handa, H.; Asano, T.; Tamura, A.; Yonekawa, Y.; Ono, H.; Tachibana, N.; Hanaoka, K. Clinical study of OKY-046, a thromboxane synthetase inhibitor, in prevention of cerebral vasospasms and delayed cerebral ischemic symptomes after subarachnoid haemorrhage due to aneurysmal rupture: A randomized double-blind study. Neurol. Res. 1989, 11, 79–88. [Google Scholar] [CrossRef] [PubMed]
  35. Tseng, M.Y.; Czosnyka, M.; Richards, H.; Pickard, J.D.; Kirkpatrick, P.J. Faculty Opinions recommendation of Effects of acute treatment with pravastatin on cerebral vasospasm, autoregulation, and delayed ischemic deficits after aneurysmal subarachnoid hemorrhage: A phase II randomized placebo-controlled trial. Stroke 2015, 36, 1627–1632. [Google Scholar] [CrossRef]
  36. Van den Bergh, W.M.; lgra, A.; Dorhout Mees, S.M.; van Kooten, F.; Dirven, C.M.; van Gijn, J.; Vermeulen, M.; Rinkel, G.J.; MASH Study Group. Randomized controlled trial of acetylsalicylic acid in aneurysmal subarachnoid hemorrhage: The MASH study. Stroke 2006, 37, 2326–2330. [Google Scholar] [CrossRef] [Green Version]
  37. Vergouwen, M.D.I.; Meijers, J.C.M.; Geskus, R.B.; Coert, B.A.; Horn, J.; Stroes, E.S.G.; Van Der Poll, T.; Vermeulen, M.; Roos, Y.B.W.E.M. Biologic Effects of Simvastatin in Patients with Aneurysmal Subarachnoid Hemorrhage: A Double-Blind, Placebo-Controlled Randomized Trial. J. Cereb. Blood Flow Metab. 2009, 29, 1444–1453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Woo, P.Y.M.; Ho, J.W.K.; Ko, N.M.W.; Li, R.P.T.; Jian, L.; Chu, A.C.H.; Kwan, M.C.L.; Chan, Y.; Wong, A.K.S.; Wong, H.-T.; et al. Randomized, placebo-controlled, double-blind, pilot trial to investigate safety and efficacy of Cerebrolysin in patients with aneurysmal subarachnoid hemorrhage. BMC Neurol. 2020, 20, 401. [Google Scholar] [CrossRef]
  39. Chen, J.; Li, M.; Zhu, X.; Chen, L.; Yang, S.; Zhang, C.; Wu, T.; Feng, X.; Wang, Y.; Chen, Q. Atorvastatin reduces cerebral vasospasm and infarction after aneurysmal subarachnoid hemorrhage in elderly Chinese adults. Aging 2020, 12, 2939–2951. [Google Scholar] [CrossRef]
  40. Fei, L.; Golwa, F. Topical application of dexamethasone to prevent cerebral vasospasm after aneurysmal subarachnoid haemorrhage: A pilot study. Clin. Drug Investig. 2007, 27, 827–832. [Google Scholar] [CrossRef]
  41. Galea, J.; Ogungbenro, K.; Hulme, S.; Patel, H.; Scarth, S.; Hoadley, M.; Illingworth, K.; McMahon, C.J.; Tzerakis, N.; King, A.; et al. Reduction of inflammation after administration of interleukin-1 receptor antagonist following aneurysmal subarachnoid hemorrhage: Results of the Subcutaneous Interleukin-1Ra in SAH (SCIL-SAH) study. J. Neurosurg. 2018, 128, 515–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Hasan, D.; Lindsay, K.W.; Wijdicks, E.F.; Murray, G.D.; Brouwers, P.J.; Bakker, W.H.; van Gijn, J.; Vermeulen, M. Effect of fludrocortisone acetate in patients with subarachnoid hemorrhage. Stroke 1989, 20, 1156–1161. [Google Scholar] [CrossRef] [Green Version]
  43. Hashi, K.; Takakura, K.; Sano, K.; Ohta, T.; Saito, I.; Okada, K. Intravenous hydrocortisone in large doses in the treatment of delayed ischemic neurological deficits following subarachnoid hemorrhage—Results of a multi-center controlled double-blind clinical study. No Shinkei= Brain Nerve 1988, 40, 373–382. [Google Scholar]
  44. Jaschinski, U.; Scherer, K.; Lichtwarck, M.; Forst, H. Impact of treatment with pravastatin on delayed ischemic disease and mortality after aneurysmal subarachnoid hemorrhage. Crit. Care 2008, 12, P112. [Google Scholar] [CrossRef] [Green Version]
  45. Lynch, J.R.; Wang, H.; McGirt, M.J.; Floyd, J.; Friedman, A.H.; Coon, A.L.; Blessing, R.; Alexander, M.J.; Graffagnino, C.; Warner, D.S.; et al. Simvastatin reduces vasospasm after aneurysmal subarachnoid hemorrhage: Results of a pilot randomized clinical trial. Stroke 2005, 36, 2024–2026. [Google Scholar] [CrossRef]
  46. Macedo, S.; Bello, Y.; Silva, A.; Siqueira, C.; Siqueira, S.; Brito, L. Effects of simvastatin in prevention of vasospasm in nontraumatic subarachnoid hemorrhage: Preliminary data. Crit. Care 2009, 13, P103. [Google Scholar] [CrossRef] [Green Version]
  47. Mori, T.; Katayama, Y.; Kawamata, T.; Hirayama, T.; Khan, M.I.; Dellinger, R.P.; Waguespack, S.G.; Shah, K.; Turgeon, R.D.; Gooderham, P.A.; et al. Improved efficiency of hypervolemic therapy with inhibition of natriuresis by fludrocortisone in patients with aneurysmal subarachnoid hemorrhage. J. Neurosurg. 1999, 91, 947–952. [Google Scholar] [CrossRef]
  48. Moro, N.; Katayama, Y.; Kojima, J.; Mori, T.; Kawamata, T. Prophylactic management of excessive natriuresis with hydrocortisone for efficient hypervolemic theerapy after subarachnoid hemorrhage. Stroke 2003, 34, 2807–2811. [Google Scholar] [CrossRef] [Green Version]
  49. Naraoka, M.; Matsuda, N.; Shimamura, N.; Asano, K.; Akasaka, K.; Takemura, A.; Hasegawa, S.; Ohkuma, H. Long-acting statin for aneurysmal subarachnoid hemorrhage: A randomized, double-blind, placebo-controlled trial. J. Cereb. Blood Flow Metab. 2017, 38, 1190–1198. [Google Scholar] [CrossRef] [PubMed]
  50. Singh, N.; Hopkins, S.J.; Hulme, S.; Galea, J.P.; Hoadley, M.; Vail, A.; Hutchinson, P.J.; Grainger, S.; Rothwell, N.J.; King, A.T.; et al. The effect of intravenous interleukin-1 receptor antagonist on inflammatory mediators in cerebrospinal fluid after subrachnoid haemorrhage: A phase II randomised controlled trial. J. Neuroinflamm. 2014, 11, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Wong, G.K.; Chan, D.Y.; Siu, D.Y.; Zee, B.C.; Poon, W.S.; Chan, M.T.; Gin, T.; Leung, M.; HDS-SAH Investigators. High-dose simvastatin for aneurysmal subarachnoid hemorrhage: Multicenter randomized controlled double-blinded clinical trial. Stroke 2015, 46, 382–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Pickard, J.D.; Murray, G.D.; Illingworth, R.; Shaw, M.D.; Teasdale, G.M.; Foy, P.M.; Humphrey, P.R.; Lang, D.A.; Nelson, R.; Richards, P. Effect of oral nimodipine on cerebral infarction and outcome after subarachnoid haemorrhage: British aneurysm nimodipine trial. BMJ 1989, 298, 636–642. [Google Scholar] [CrossRef] [Green Version]
  53. Rouzer, C.A.; Marnett, L.J. Structural and Chemical Biology of the Interaction of Cyclooxygenase with Substrates and Non-Steroidal Anti-Inflammory Drugs. Chem. Rev. 2020, 120, 7592–7641. [Google Scholar] [CrossRef]
  54. Wu, F.; Liu, Z.; Li, G.; Zhou, L.; Huang, K.; Wu, Z.; Zhan, R.; Shen, J. Inflammation and Oxidative Stress: Potential Targets for Improving Prognosis After Subarachnoid Hemorrhage. Front. Cell. Neurosci. 2021, 15, 739506. [Google Scholar] [CrossRef]
  55. Dinh, Y.R.T.; Jomaa, A.; Callebert, J.; Reynier-Rebuffel, A.-M.; Tedgui, A.; Savarit, A.; Sercombe, R. Overexpression of Cyclooxygenase-2 in Rabbit Basilar Artery Endothelial Cells after Subarachnoid Hemorrhage. Neurosurgery 2001, 48, 626–635. [Google Scholar] [CrossRef] [PubMed]
  56. Ayer, R.; Jadhav, V.; Sugawara, T.; Zhang, J.H. The Neuroprotective Effects of Cyclooxygenase-2 Inhibition in a Mouse Model of Aneurysmal Subarachnoid Hemorrhage. Acta Neurochir. Suppl. 2011, 111, 145–149. [Google Scholar] [CrossRef]
  57. Roumie, C.L.; Mitchel, E.F., Jr.; Kaltenbach, L.; Arbogast, P.G.; Gideon, P.; Griffin, M.R. Nonaspirin NSAIDs, cyclooxygenase 2 inhibitors, and the risk for stroke. Stroke 2008, 39, 2037–2045. [Google Scholar] [CrossRef] [Green Version]
  58. Parkhutik, V.; Lago, A.; Tembl, J.I.; Rubio, C.; Fuset, M.P.; Vallés, J.; Santos, M.T.; Moscardo, A. Influence of COX-inhibiting Analgesics on the Platelet Function of Patients with Subarachnoid Hemorrhage. J. Stroke Cerebrovasc. Dis. 2012, 21, 755–759. [Google Scholar] [CrossRef] [PubMed]
  59. Park, H.K.; Lee, S.H.; Chu, K.; Roh, J.K. Effects of celecoxib on volumes of hematoma and edema in patients with primary intracerebral hemorrhhage. J. Neurol. Sci. 2009, 279, 43–46. [Google Scholar] [CrossRef] [PubMed]
  60. Caldwell, B.; Aldington, S.; Weatherall, M.; Shirtcliffe, P.; Beasley, R. Risk of Cardiovascular Events and Celecoxib: A Systematic Review and Meta-Analysis. J. R. Soc. Med. 2006, 99, 132–140. [Google Scholar] [CrossRef] [Green Version]
  61. Coyle, A.T.; Miggin, S.M.; Kinsella, B.T. Characterization of the 5′ untranslated region of alpha and beta isoforms of the human thromboxane A2 receptor (TP). Differential promoter utilization by the TP isoforms. Eur. J. Biochem. 2002, 269, 4058–4073. [Google Scholar] [CrossRef]
  62. Davì, G.; Patrono, C. Platelet Activation and Atherothrombosis. N. Engl. J. Med. 2007, 357, 2482–2494. [Google Scholar] [CrossRef]
  63. Ansar, S.; Larsen, C.; Maddahi, A.; Edvinsson, L. Subarachnoid hemorrhage induces enhanced expression of thromboxane A2 receptors in rat cerebral arteries. Brain Res. 2010, 1316, 163–172. [Google Scholar] [CrossRef] [PubMed]
  64. Tohgi, H.; Konno, S.; Tamura, K.; Kimura, B.; Kawano, K. Effects of low-to-high doses of aspirin on platelet aggregability and metabolites of thromboxane A2 and prostacyclin. Stroke 1992, 23, 1400–1403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Vinge, E.; Brandt, L.; Ljunggren, B.; Andersson, K.E. Thromboxane B2 levels in serum during continous administration of nimodipine to patients with aneurysmal subarachnoid hemorrhage. Stroke 1988, 19, 644–647. [Google Scholar] [CrossRef] [Green Version]
  66. Collins, T.; Read, M.A.; Neish, A.S.; Whitley, M.Z.; Thanos, D.; Maniatis, T. Transcriptional regulation of endothelial cell adhesion molecules: NF-kappa B and cytokine-inducible enhancers. FASEB J. 1995, 9, 899–909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Node, K.; Huo, Y.; Ruan, X.; Yang, B.; Spiecker, M.; Ley, K.; Zeldin, D.C.; Liao, J.K. Anti-inflammatory Properties of Cytochrome P450 Epoxygenase-Derived Eicosanoids. Science 1999, 285, 1276–1279. [Google Scholar] [CrossRef] [Green Version]
  68. Xiao, L.; Liu, Y.; Wang, N. New paradigms in inflammatory signaling in vascular endothelial cells. Am. J. Physiol. Circ. Physiol. 2014, 306, H317–H325. [Google Scholar] [CrossRef]
  69. Muldoon, L.L.; Alvarez, J.I.; Begley, D.J.; Boado, R.J.; Del Zoppo, G.J.; Doolittle, N.D.; Engelhardt, B.; Hallenbeck, J.M.; Lonser, R.R.; Ohlfest, J.R.; et al. Immunologic Privilege in the Central Nervous System and the Blood–Brain Barrier. J. Cereb. Blood Flow Metab. 2013, 33, 13–21. [Google Scholar] [CrossRef] [Green Version]
  70. Siler, D.A.; Berlow, Y.A.; Kukino, A.; Davis, C.M.; Nelson, J.W.; Grafe, M.R.; Ono, H.; Cetas, J.S.; Pike, M.; Alkayed, N.J. Soluble Epoxide Hydrolase in Hydrocephalus, Cerebral Edema, and Vascular Inflammation after Subarachnoid Hemorrhage. Stroke 2015, 46, 1916–1922. [Google Scholar] [CrossRef] [Green Version]
  71. Greenwood, J.; Etienne-Manneville, S.; Adamson, P.; Couraud, P.-O. Lymphocyte migration into the central nervous system: Implication of ICAM-1 signalling at the blood–brain barrier. Vasc. Pharmacol. 2002, 38, 315–322. [Google Scholar] [CrossRef] [PubMed]
  72. Laufs, U.; La Fata, V.; Plutzky, J.; Liao, J.K. Upregulation of Endothelial Nitric Oxide Synthase by HMG CoA Reductase Inhibitors. Circulation 1998, 97, 1129–1135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. McGirt, M.J.; Lynch, J.R.; Parra, A.; Sheng, H.; Pearlstein, R.D.; Laskowitz, D.T.; Pelligrino, D.A.; Warner, D.S. Simvastatin Increases Endothelial Nitric Oxide Synthase and Ameliorates Cerebral Vasospasm Resulting From Subarachnoid Hemorrhage. Stroke 2002, 33, 2950–2956. [Google Scholar] [CrossRef]
  74. Weitz-Schmidt, G. Statins as anti-inflammatory agents. Trends Pharmacol. Sci. 2002, 23, 482–487. [Google Scholar] [CrossRef]
  75. McGirt, M.J.; Pradilla, G.; Legnani, F.G.; Thai, Q.-A.; Recinos, P.F.; Tamargo, R.J.; Clatterbuck, R.E. Systemic Administration of Simvastatin after the Onset of Experimental Subarachnoid Hemorrhage Attenuates Cerebral Vasospasm. Neurosurgery 2006, 58, 945–951. [Google Scholar] [CrossRef]
  76. Vergouwen, M.D.; de Haan, R.J.; Vermeulen, M.; Roos, Y.B. Effect of statin treatment on vasospasm, delayed cerebral ischemia, and functional outcome in patients with aneurysmal subarachnoid hemorrhage: A systematic review and meta-analysis update. Stroke 2010, 41, e47–e52. [Google Scholar] [CrossRef] [Green Version]
  77. Liu, T.; Zhong, S.; Zhai, Q.; Zhang, X.; Jing, H.; Li, K.; Liu, S.; Han, S.; Li, L.; Shi, X.; et al. Optimal Course of Statins for Patients With Aneurysmal Subarachnoid Hemorrhage: Is Longer Treatment Better? A Meta-Analysis of Randomized Controlled Trials. Front. Neurosci. 2021, 15, 757505. [Google Scholar] [CrossRef] [PubMed]
  78. Maher, M.; Schweizer, T.A.; Macdonald, R.L. Treatment of Spontaneous Subarachnoid Hemorrhage: Guidelines and Gaps. Stroke 2020, 51, 1326–1332. [Google Scholar] [CrossRef] [PubMed]
  79. Guan, X.; Wang, Y.; Kai, G.; Zhao, S.; Huang, T.; Li, Y.; Xu, Y.; Zhang, L.; Pang, T. Cerebrolysin Ameliorates Focal Cerebral Ischemia Injury Through Neuroinflammatory Inhibition via CREB/PGC-1α Pathway. Front. Pharmacol. 2019, 10, 1245. [Google Scholar] [CrossRef]
  80. Muresanu, D.F.; Heiss, W.D.; Hoemberg, V.; Bajenaru, O.; Popescu, C.D.; Vester, J.C.; Rahlfs, V.W.; Doppler, E.; Meier, D.; Moessler, H.; et al. Cerebrolysin and recovery after stroke (CARS): A randomized, placebo-controlled, double-blind, multicenter trial. Stroke 2016, 47, 151–159. [Google Scholar] [CrossRef]
  81. Xue, L.X.; Zhang, T.; Zhao, Y.W.; Geng, Z.; Chen, J.J.; Chen, H. Efficacy and safety comparison of DL-3-n.butylphtalide and Cerebrolysin: Effects on neurological and behavioral outcomes in acute ischemic stroke. Exp. Ther. Med. 2016, 11, 2015–2020. [Google Scholar] [CrossRef] [Green Version]
  82. Lang, W.; Stadler, C.H.; Poljakovic, Z.; Fleet, D. The Lyse Study Group. A Prospective, Randomized, Placebo-Controlled, Double-Blind Trial about Safety and Efficacy of Combined Treatment with Alteplase (rt-PA) and Cerebrolysin in Acute Ischaemic Hemispheric Stroke. Int. J. Stroke 2012, 8, 95–104. [Google Scholar] [CrossRef]
  83. Chang, W.H.; Park, C.H.; Kim, D.Y.; Shin, Y.I.; Ko, M.H.; Lee, A.; Jang, S.Y.; Kim, Y.H. Cerebrolysin combined with rehabilitation promotes motro recovery in patients with severe motor impairment after stroke. BMC Neurol. 2016, 16, 31. [Google Scholar] [CrossRef] [Green Version]
  84. Gharagozli, K.; Harandi, A.; Houshmand, S.; Akbari, N.; Muresanu, D.; Vester, J.; Winter, S.; Moessler, H. Efficacy and safety of Cerebrolysin treatment in early recovery after acute ischemic stroke: A randomized, placebo-controlled, double-blinded, multicenter clinical trial. J. Med. Life 2017, 10, 153–160. [Google Scholar]
  85. Rezaei, Y.; Amiri-Nikpour, M.R.; Nazarbaghi, S.; Ahmadi-Salmasi, B.; Mokari, T.; Tahmtan, O. Cerebrolysin effects on neurological outcomes and cerebral blood flow in acute ischemic stroke. Neuropsychiatr. Dis. Treat. 2014, 10, 2299–2306. [Google Scholar] [CrossRef] [Green Version]
  86. Park, Y.K.; Yi, H.-J.; Choi, K.-S.; Lee, Y.-J.; Kim, D.-W.; Kwon, S.M. Cerebrolysin for the Treatment of Aneurysmal Subarachnoid Hemorrhage in Adults: A Retrospective Chart Review. Adv. Ther. 2018, 35, 2224–2235. [Google Scholar] [CrossRef] [PubMed]
  87. Rıos, C.; Nader-Kawachi, J.; Rodriguez-Payán, A.J.; Nava-Ruiz, C. Neuroprotective effect of dapsone in an occlusive model of focal ischemia in rats. Brain Res. 2004, 999, 212–215. [Google Scholar] [CrossRef] [PubMed]
  88. Suda, T.; Suzuki, Y.; Matsui, T.; Inoue, T.; Niide, O.; Yoshimaru, T.; Suzuki, H.; Ra, C.; Ochiai, T. Dapsone suppresses human neutrophil superoxide production and elastase release in a calcium-dependent manner. Br. J. Dermatol. 2005, 152, 887–895. [Google Scholar] [CrossRef]
  89. He, J.; Zhang, X.; He, W.; Xie, Y.; Chen, Y.; Yang, Y.; Chen, R. Neuroprotective effects of zonisamide on cerebral ischemia injury via inhibition of neuronal apoptosis. Braz. J. Med. Biol. Res. 2021, 54, e10498. [Google Scholar] [CrossRef] [PubMed]
  90. Nader-Kawachi, J.; Góngora-Rivera, F.; Santos-Zambrano, J.; Calzada, P.; Ríos, C. Neuroprotective effect of dapsone in patients with acute ischemic stroke: A pilot study. Neurol. Res. 2007, 29, 331–334. [Google Scholar] [CrossRef]
  91. Mistry, A.; Mistry, E.A.; Kumar, N.G.; Froehler, M.T.; Fusco, M.R.; Chitale, R.V. Corticosteroids in the Management of Hyponatremia, Hypovolemia, and Vasospasm in Subarachnoid Hemorrhage: A Meta-Analysis. Cerebrovasc. Dis. 2016, 42, 263–271. [Google Scholar] [CrossRef]
  92. Brown, R.J.; Epling, B.P.; Staff, I.; Fortunato, G.; Grady, J.J.; McCullough, L.D. Polyuria and cerebral vasospasm after aneurysmal subarachnoid hemorrhage. BMC Neurol. 2015, 15, 201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Egge, A.; Waterloo, K.; Sjoholm, H.; Solberg, T.; Ingebrigtsen, T.; Romner, B. Prophylactic hyperdynamic postoperative fluid therapy after aneurysmal subarachnoid hemorrhage: A clinical, prospective, randomized, controlled study. Neurosurgery 2001, 49, 593–605. [Google Scholar] [PubMed] [Green Version]
  94. Lennihan, L.; Mayer, S.A.; Fink, M.E.; Beckford, A.; Paik, M.C.; Zhang, H.; Wu, Y.C.; Klebanoff, L.M.; Raps, E.C.; Solomon, R.A. Effect of hypervolemic therapy on cerebral blood flow after subarachnoid hemorrhage: A randomized controlled trial. Stroke 2000, 31, 383–391. [Google Scholar] [CrossRef] [PubMed]
  95. Nakagawa, I.; Hironaka, Y.; Nishimura, F.; Takeshima, Y.; Matsuda, R.; Yamada, S.; Motoyama, Y.; Park, Y.-S.; Nakase, H. Early Inhibition of Natriuresis Suppresses Symptomatic Cerebral Vasospasm in Patients with Aneurysmal Subarachnoid Hemorrhage. Cerebrovasc. Dis. 2013, 35, 131–137. [Google Scholar] [CrossRef]
  96. Miller, B.A.; Turan, N.; Chau, M.; Pradilla, G. Inflammation, vasospasm, and brain injury after subarachnoid hemorrhage. Biomed. Res. Int. 2014, 2014, 384342. [Google Scholar] [CrossRef] [Green Version]
  97. Chaudhry, S.R.; Stoffel-Wagner, B.; Kinfe, T.M.; Güresir, E.; Vatter, H.; Dietrich, D.; Lamprecht, A.; Muhammad, S. Elevated Systemic IL-6 Levels in Patients with Aneurysmal Subarachnoid Hemorrhage Is an Unspecific Marker for Post-SAH Complications. Int. J. Mol. Sci. 2017, 18, 2580. [Google Scholar] [CrossRef] [Green Version]
  98. Chyatte, D.; Fode, N.C.R.N.; Nichols, D.A.; Sundt, T.M. Preliminary Report: Effects of High Dose Methylprednisolone on Delayed Cerebral Ischemia in Patients at High Risk for Vasospasm after Aneurysmal Subarachnoid Hemorrhage. Neurosurgery 1987, 21, 157–160. [Google Scholar] [CrossRef]
  99. McGirt, M.J.; Mavropoulos, J.C.; McGirt, L.Y.; Alexander, M.J.; Friedman, A.H.; Laskowitz, D.T.; Lynch, J.R. Leukcytosis as an independent risk factor for cerebral vasospasm following aneurysmal subarachnoid hemorrhage. J. Neurosurg. 2003, 98, 1222–1226. [Google Scholar] [CrossRef] [Green Version]
  100. Schürkämper, M.; Medele, R.; Zausinger, S.; Schmid-Elsaesser, R.; Steiger, H.-J. Dexamethasone in the treatment of subarachnoid hemorrhage revisited: A comparative analysis of the effect of the total dose on complications and outcome. J. Clin. Neurosci. 2004, 11, 20–24. [Google Scholar] [CrossRef]
  101. Güresir, E.; Lampmann, T.; Bele, S.; Czabanka, M.; Czorlich, P.; Gempt, J.; Goldbrunner, R.; Hurth, H.; Hermann, E.; Jabbarli, R.; et al. Fight INflammation to Improve outcome after aneurysmal Subarachnoid HEmorRhage (FINISHER) trial: Study protocol for a randomized controlled trial. Int. J. Stroke 2022, 18, 242–247. [Google Scholar] [CrossRef]
  102. Brotman, D.J.; Girod, J.P.; Posch, A.; Jani, J.T.; Patel, J.V.; Gupta, M.; Lip, G.Y.; Reddy, S.; Kickler, T.S. Effects of short-term glucocorticoids on hemostatic factors in healthy volunteers. Thromb. Res. 2006, 118, 247–252. [Google Scholar] [CrossRef]
  103. Isidori, A.M.; Minnetti, M.; Sbardella, E.; Graziadio, C.; Grossman, A.B. Mechanisms in endocrinology: The spectrum of haemostatic abnormalities in glucocorticoid excess and defect. Eur. J. Endocrinol. 2015, 173, R101–R113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Ornelas, A.; Zacharias-Millward, N.; Menter, D.G.; Davis, J.S.; Lichtenberger, L.; Hawke, D.; Hawk, E.; Vilar, E.; Bhattacharya, P.; Millward, S. Beyond COX-1: The effects of aspirin on platelet biology and potential mechanisms of chemoprevention. Cancer Metastasis Rev. 2017, 36, 289–303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA flow chart showing the study selection of the present meta-analysis.
Figure 1. PRISMA flow chart showing the study selection of the present meta-analysis.
Jcm 12 04165 g001
Figure 2. (A) Risk of bias evaluation for each kind of bias. (B) Summary of risk of bias of the individual randomized controlled trials (reviewers’ judgments about each risk of bias characteristic of the included studies: “+” constitutes low risk; “?” constitutes unclear risk).
Figure 2. (A) Risk of bias evaluation for each kind of bias. (B) Summary of risk of bias of the individual randomized controlled trials (reviewers’ judgments about each risk of bias characteristic of the included studies: “+” constitutes low risk; “?” constitutes unclear risk).
Jcm 12 04165 g002
Figure 3. Forest Plots displaying OR and 95% CI estimates for death in studies evaluating anti-inflammatory therapies compared to conventional therapy in aSAH [25,26,27,28,29,30,32,33,34,35,36,37,38]. Squares constitute the odds ratio; the bigger the square, the greater the weight given because of the narrower 95% CI. Diamond shows the odds ratio of the overall data.
Figure 3. Forest Plots displaying OR and 95% CI estimates for death in studies evaluating anti-inflammatory therapies compared to conventional therapy in aSAH [25,26,27,28,29,30,32,33,34,35,36,37,38]. Squares constitute the odds ratio; the bigger the square, the greater the weight given because of the narrower 95% CI. Diamond shows the odds ratio of the overall data.
Jcm 12 04165 g003
Figure 4. Forest Plots displaying OR and 95% CI estimates for a good neurological outcome (using mRS grading: mRS ≤ 2) in studies evaluating anti-inflammatory therapies compared to placebo therapy in aneurysmal SAH [25,26,27,29,30,31,32,35,38]. Squares constitute the odds ratio; the bigger the square, the greater the weight given because of the narrower 95% CI. Diamond shows the odds ratio of the overall data.
Figure 4. Forest Plots displaying OR and 95% CI estimates for a good neurological outcome (using mRS grading: mRS ≤ 2) in studies evaluating anti-inflammatory therapies compared to placebo therapy in aneurysmal SAH [25,26,27,29,30,31,32,35,38]. Squares constitute the odds ratio; the bigger the square, the greater the weight given because of the narrower 95% CI. Diamond shows the odds ratio of the overall data.
Jcm 12 04165 g004
Figure 5. (A) Funnel plot visualizing the analysis of publications bias for the endpoint survival in 13 studies [25,26,27,28,29,30,32,33,34,35,36,37,38]. (B) Funnel plot visualizing the analysis of publications bias for the endpoint neurological outcome in nine studies [25,26,27,29,30,31,32,35,38].
Figure 5. (A) Funnel plot visualizing the analysis of publications bias for the endpoint survival in 13 studies [25,26,27,28,29,30,32,33,34,35,36,37,38]. (B) Funnel plot visualizing the analysis of publications bias for the endpoint neurological outcome in nine studies [25,26,27,29,30,31,32,35,38].
Jcm 12 04165 g005
Table 1. Major characteristics of anti-inflammatory studies included in the present meta-analysis.
Table 1. Major characteristics of anti-inflammatory studies included in the present meta-analysis.
NameYearTreatments and Sample SizeDosesTreatment DurationRecruiting Area
Chou et al. [25]2008Simvastatin = 19 versus SNT = 2080 mg oral dailyUntil discharge from NICU or until 21 daysUnited States of America
Diringer et al. [26]2016Simvastatin = 12 versus SNT = 1280 mg oral daily21 daysUnited States of America
Garcia-Pastor et al. [27]2022Dapsone = 26 versus SNT = 22100 mg oral dailyWithin 5 days after ictus until day 15Mexico
Garg et al. [28]2013Simvastatin = 19 versus SNT = 1980 mg daily for 14 days14 daysIndia
Gomis et al. [29]2010Methylprednisolone = 49 versus SNT = 4616 mg/kg intravenous daily3 daysFrance
Hop et al. [30]2000ASA = 24 versus SNT = 26100 mg suppositories daily21 daysNetherlands
Katayama et al. [31]2007Hydrocortisone = 35 versus SNT = 361200 mg/d (day 0–10), 600 mg/d (day 11–12), 300 mg/d (day 13–14) intravenously14 daysJapan
Kirkpatrick et al. [32]2014Simvastatin = 391 versus SNT = 41240 mg oral daily3 weeksUK, Canada, Colombia, Italy, Russia, Singapore, Sweden, Uruguay, USA
Martini et al. [33]2022Epoxide hydrolase inhibitor = 10 versus SNT = 1010 mg oral daily10 daysUnited States of America
Suzuki et al. [34]1989OKY-046 = 172 versus SNT = 8680 mg/day (low-dose group)
400 mg/day (high-dose group) intravenously
10 to 14 daysJapan
Tseng et al. [35]2005Pravastatin = 40 versus SNT = 4040 mg oral dailyWithin 3 days after ictus for 14 days or until dischargeUnited Kingdom
Van den Bergh et al. [36]2006ASA = 83 versus SNT = 70100 mg suppositories daily14 daysNetherlands
Vergouwen et al. [37]2009Simvastatin = 14 versus SNT = 1680 mg oral dailyWithin 3 days after SAH until day 14Netherlands
Woo et al. [38]2020Cerebrolysin = 25 versus SNT = 2530 mL daily for 14 days14 daysHong Kong
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wach, J.; Vychopen, M.; Güresir, A.; Güresir, E. Anti-Inflammatory Drug Therapy in Aneurysmal Subarachnoid Hemorrhage: A Systematic Review and Meta-Analysis of Prospective Randomized and Placebo-Controlled Trials. J. Clin. Med. 2023, 12, 4165. https://doi.org/10.3390/jcm12124165

AMA Style

Wach J, Vychopen M, Güresir A, Güresir E. Anti-Inflammatory Drug Therapy in Aneurysmal Subarachnoid Hemorrhage: A Systematic Review and Meta-Analysis of Prospective Randomized and Placebo-Controlled Trials. Journal of Clinical Medicine. 2023; 12(12):4165. https://doi.org/10.3390/jcm12124165

Chicago/Turabian Style

Wach, Johannes, Martin Vychopen, Agi Güresir, and Erdem Güresir. 2023. "Anti-Inflammatory Drug Therapy in Aneurysmal Subarachnoid Hemorrhage: A Systematic Review and Meta-Analysis of Prospective Randomized and Placebo-Controlled Trials" Journal of Clinical Medicine 12, no. 12: 4165. https://doi.org/10.3390/jcm12124165

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop