Efficacy of Minocycline in Acute Ischemic Stroke: A Systematic Review and Meta-Analysis of Rodent and Clinical Studies

Zhaofu Sheng, Yang Liu, Hongmin Li, Wei Zheng, Bin Xia, Xin Zhang, V Wee Yong, Mengzhou Xue, Zhaofu Sheng, Yang Liu, Hongmin Li, Wei Zheng, Bin Xia, Xin Zhang, V Wee Yong, Mengzhou Xue

Abstract

Objectives: This study aimed to assess the efficacy of minocycline for the treatment of acute ischemic stroke. Background: While there have been meta-analysis that surveyed the efficacy of minocycline in the treatment of acute stroke, they have some methodological limitations. We performed a new systematic review which was distinct from previous one by adding new outcomes and including new studies. Methods: Document retrieval was executed through PubMed, Cochrane Central Register of Controlled Trials, the Stroke Center, NIH's Clinical Trials, Current Controlled Trials, and the WHO International Clinical Trials Registry Platform Search Portal before Jan 2018. The data meeting the inclusion criteria were extracted. Before meta-analysis, publication bias and heterogeneity of included studies were surveyed. Random and fixed-effects models were employed to calculate pooled estimates and 95% confidence intervals (CIs). Additionally, sensitivity and subgroup analyses were implemented. Result: For clinical studies, 4 trials with 201 patients in the minocycline group, and 195 patients in the control group met the inclusion criteria; 3 were randomized trials. At the end of 90-day follow up or discharge day, results showed that the groups receiving minocycline were superior to the control group, with significant differences in the NIHSS scores (mean difference [MD], -2.75; 95% CI, -4.78, 0.27; p = 0.03) and mRS scores (MD, -0.98; 95% CI, -1.27, -0.69; p < 0.01), but not Barthel Index Score (MD, 9.04; 95% CI, -0.78, 18.07; p = 0.07). For rodent experiments, 14 studies were included. Neurological severity scores (NSS) was significantly improved (MD, -1.38; 95% CI, -1.64, -1.31; p < 0.01) and infarct volume was obviously reduced (Std mean difference [SMD], -2.38; 95% CI, -3.40, -1.36; p < 0.01) in the minocycline group. Heterogeneity among the studies was proved to exist for infarct volume (Chi2 = 116.12, p < 0.01; I2 = 0.89) but not for other variables. Conclusions: Based on the results in our study, minocycline appears as an effective therapeutic option for acute ischemic stroke.

Keywords: clinical study; ischemic stroke; meta-analysis; minocycline; rodent study.

Figures

Figure 1
Figure 1
Flow chart of literature assessment for studies.
Figure 2
Figure 2
Methodological quality assessment for randomized clinical trials. (A) Risk of bias graph: review judgments about each risk of bias item presented as percentages across all included studies. (B) Risk of bias summary: review judgments about each risk of bias item for each included study. “+,” low risk of bias; “–,” high risk of bias; “?,” unclear risk of bias.
Figure 3
Figure 3
Funnel plot assessing publication bias of rodent experimental studies. (A) Neurological Severity Score, NSS. (B) Infarct volume. Vertical lines represent the summary effects; dashed lines denote the 95% CIs.
Figure 4
Figure 4
Forest plot illustrating the meta-analysis of the clinical outcome. (A) National Institutes of Health Stroke Scale, NIHSS. (B) modified Rankin Scale, mRS. (C) Barthel Index, BI.
Figure 5
Figure 5
Forest plot illustrating the meta-analysis of the rodent experimental outcome. (A) Neurological Severity Score, NSS. (B) NSS stratified by administration route. (C) Infarct volume.

References

    1. Lloyd-Jones D, Adams R, Carnethon M, De Simone G, Ferguson TB, Flegal K, et al. Heart disease and stroke statistics−2009 update: a report from the american heart association statistics committee and stroke statistics subcommittee. Circulation (2009) 119:480–6. 10.1161/CIRCULATIONAHA.108.191259
    1. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Borden WB, et al. Heart disease and stroke statistics−2013 update: A report from the american heart association. Circulation (2013) 127:e6–e245. 10.1161/CIR.0b013e31828124ad
    1. Yong VW, Wells J, Giuliani F, Casha S, Power C, Metz LM. The promise of minocycline in neurology. Lancet (2004) 3:744–51. 10.1016/S1474-4422(04)00937-8
    1. Amiri-Nikpour MR, Nazarbaghi S, Hamdi-Holasou M, Rezaei Y. An open-label evaluator-blinded clinical study of minocycline neuroprotection in ischemic stroke: Gender-dependent effect. Acta Neurol Scand. (2015) 131:45–50. 10.1111/ane.12296
    1. Switzer JA, Hess DC, Ergul A, Waller JL, Machado LS, Portik-Dobos V, et al. . Matrix metalloproteinase-9 in an exploratory trial of intravenous minocycline for acute ischemic stroke. Stroke (2011) 42:2633–5. 10.1161/STROKEAHA.111.618215
    1. Lampl Y, Boaz M, Gilad R, Lorberboym M, Dabby R, Rapoport A, et al. . Minocycline treatment in acute stroke: An open-label, evaluator-blinded study. Neurology (2007) 69:1404–10. 10.1212/01.wnl.0000277487.04281.db
    1. Kohler E, Prentice DA, Bates TR, Hankey GJ, Claxton A, van Heerden J, et al. . Intravenous minocycline in acute stroke: a randomized, controlled pilot study and meta-analysis. Stroke (2013) 44:2493–9. 10.1161/STROKEAHA.113.000780
    1. Padma Srivastava MV, Bhasin A, Bhatia R, Garg A, Gaikwad S, Prasad K, et al. Efficacy of minocycline in acute ischemic stroke: A single-blinded, placebo-controlled trial. Neurology (2012) 60:23–8. 10.4103/0028-3886.93584
    1. Fouda AY, Newsome AS, Spellicy S, Waller JL, Zhi W, Hess DC, et al. . Minocycline in acute cerebral hemorrhage: An early phase randomized trial. Stroke (2017) 48:2885–7. 10.1161/STROKEAHA.117.018658
    1. Chang JJ, Kim-Tenser M, Emanuel BA, Jones GM, Chapple K, Alikhani A, et al. . Minocycline and matrix metalloproteinase inhibition in acute intracerebral hemorrhage: a pilot study. Eur J Neurol. (2017) 24:1384–91. 10.1111/ene.13403
    1. Quinn TJ, Dawson J, Walters MR, Lees KR. Reliability of the modified rankin scale: A systematic review. Stroke (2009) 40:3393–5. 10.1161/STROKEAHA.109.557256
    1. Chaisinanunkul N, Adeoye O, Lewis RJ, Grotta JC, Broderick J, Jovin TG, et al. . Adopting a patient-centered approach to primary outcome analysis of acute stroke trials using a utility-weighted modified rankin scale. Stroke (2015) 46:2238–43. 10.1161/STROKEAHA.114.008547
    1. Malhotra K, Chang JJ, Khunger A, Blacker D, Switzer JA, Goyal N, et al. . Minocycline for acute stroke treatment: a systematic review and meta-analysis of randomized clinical trials. J Neurol. (2018) 265:1871–9. 10.1007/s00415-018-8935-3
    1. Muir KW, Weir CJ, Murray GD, Povey C, Lees KR. Comparison of neurological scales and scoring systems for acute stroke prognosis. Stroke (1996) 27:1817–20. 10.1161/01.STR.27.10.1817
    1. Cioncoloni D, Piu P, Tassi R, Acampa M, Guideri F, Taddei S, et al. . Relationship between the modified rankin scale and the barthel index in the process of functional recovery after stroke. NeuroRehabilitation (2012) 30:315–322. 10.3233/NRE-2012-0761
    1. Quinn TJ, Langhorne P, Stott DJ. Barthel index for stroke trials: development, properties, and application. Stroke (2011) 42:1146–51. 10.1161/STROKEAHA.110.598540
    1. Volmink J, Siegfried N, Robertson K, Gulmezoglu AM. Research synthesis and dissemination as a bridge to knowledge management: the cochrane collaboration. Bull World Health Organiz. (2004) 82:778–83.
    1. Slim K, Nini E, Forestier D, Kwiatkowski F, Panis Y, Chipponi J. Methodological index for non-randomized studies (minors): Development and validation of a new instrument. ANZ J Surg. (2003) 73:712–6. 10.1046/j.1445-2197.2003.02748.x
    1. Sutton AJ, Duval SJ, Tweedie RL, Abrams KR, Jones DR. Empirical assessment of effect of publication bias on meta-analyses. BMJ (2000) 320:1574–7. 10.1136/bmj.320.7249.1574
    1. Macleod MR, O'Collins T, Howells DW, Donnan GA. Pooling of animal experimental data reveals influence of study design and publication bias. Stroke (2004) 35:1203–8. 10.1161/01.STR.0000125719.25853.20
    1. Crossley NA, Sena E, Goehler J, Horn J, van der Worp B, Bath PM, et al. . Empirical evidence of bias in the design of experimental stroke studies: a metaepidemiologic approach. Stroke (2008) 39:929–34. 10.1161/STROKEAHA.107.498725
    1. Sardar P, Chatterjee S, Giri J, Kundu A, Tandar A, Sen P, et al. . Endovascular therapy for acute ischaemic stroke: a systematic review and meta-analysis of randomized trials. Eur Heart J. (2015) 36:2373–80. 10.1093/eurheartj/ehv270
    1. Lambrinos A, Schaink AK, Dhalla I, Krings T, Casaubon LK, Sikich N, et al. . Mechanical thrombectomy in acute ischemic stroke: a systematic review. Can J Neurol Sci. (2016) 43:455–60. 10.1017/cjn.2016.30
    1. Bhatt LK, Addepalli V. Potentiation of aspirin-induced cerebroprotection by minocycline: a therapeutic approach to attenuate exacerbation of transient focal cerebral ischaemia. Diabetes Vasc Dis Res. (2012) 9:25–34. 10.1177/1479164111427753
    1. Hayakawa K, Mishima K, Nozako M, Hazekawa M, Mishima S, Fujioka M, et al. . Delayed treatment with minocycline ameliorates neurologic impairment through activated microglia expressing a high-mobility group box1-inhibiting mechanism. Stroke (2008) 39:951–8. 10.1161/STROKEAHA.107.495820
    1. Hoda MN, Li W, Ahmad A, Ogbi S, Zemskova MA, Johnson MH, et al. . Sex-independent neuroprotection with minocycline after experimental thromboembolic stroke. Exp Trans Stroke Med. (2011) 3:16. 10.1186/2040-7378-3-16
    1. Jin X, Liu J, Liu KJ, Rosenberg GA, Yang Y, Liu W. Normobaric hyperoxia combined with minocycline provides greater neuroprotection than either alone in transient focal cerebral ischemia. Exp Neurol. (2013) 240:9–16. 10.1016/j.expneurol.2012.11.018
    1. Jin Z, Liang J, Wang J, Kolattukudy PE. Mcp-induced protein 1 mediates the minocycline-induced neuroprotection against cerebral ischemia/reperfusion injury in vitro and in vivo. J Neuroinflamm. (2015) 12:39. 10.1186/s12974-015-0264-1
    1. Li J, McCullough LD. Sex differences in minocycline-induced neuroprotection after experimental stroke. J Cereb Blood Flow Metab. (2009) 29:670–4. 10.1038/jcbfm.2009.3
    1. Liu Z, Fan Y, Won SJ, Neumann M, Hu D, Zhou L, et al. . Chronic treatment with minocycline preserves adult new neurons and reduces functional impairment after focal cerebral ischemia. Stroke (2007) 38:146–52. 10.1161/
    1. Martin A, Boisgard R, Kassiou M, Dolle F, Tavitian B. Reduced pbr/tspo expression after minocycline treatment in a rat model of focal cerebral ischemia: a pet study using [(18)f]dpa-714. Mol Imag Biol. (2011) 13:10–5. 10.1007/s11307-010-0324-y
    1. Matsukawa N, Yasuhara T, Hara K, Xu L, Maki M, Yu G, et al. . Therapeutic targets and limits of minocycline neuroprotection in experimental ischemic stroke. BMC Neurosci. (2009) 10:126. 10.1186/1471-2202-10-126
    1. Murata Y, Rosell A, Scannevin RH, Rhodes KJ, Wang X, Lo EH. Extension of the thrombolytic time window with minocycline in experimental stroke. Stroke (2008) 39:3372–7. 10.1161/STROKEAHA.108.514026
    1. Soliman S, Ishrat T, Fouda AY, Patel A, Pillai B, Fagan SC. Sequential therapy with minocycline and candesartan improves long-term recovery after experimental stroke. Transl Stroke Res. (2015) 6:309–22. 10.1007/s12975-015-0408-8
    1. Wang CX, Yang T, Noor R, Shuaib A. Delayed minocycline but not delayed mild hypothermia protects against embolic stroke. BMC Neurol. (2002) 2:2 10.1186/1471-2377-2-2
    1. Xu L, Fagan SC, Waller JL, Edwards D, Borlongan CV, Zheng J, et al. . Low dose intravenous minocycline is neuroprotective after middle cerebral artery occlusion-reperfusion in rats. BMC Neurol. (2004) 4:7. 10.1186/1471-2377-4-7
    1. Yenari MA, Xu L, Tang XN, Qiao Y, Giffard RG. Microglia potentiate damage to blood-brain barrier constituents: Improvement by minocycline in vivo and in vitro. Stroke (2006) 37:1087–93. 10.1161/
    1. Xue M, Mikliaeva EI, Casha S, Zygun D, Demchuk A, Yong VW. Improving outcomes of neuroprotection by minocycline: guides from cell culture and intracerebral hemorrhage in mice. Am J Pathol. (2010) 176:1193–202. 10.2353/ajpath.2010.090361
    1. Katsuki H. Exploring neuroprotective drug therapies for intracerebral hemorrhage. J Pharmacol Sci. (2010) 114:366–78. 10.1254/jphs.10R05CR
    1. Kikuchi K, Uchikado H, Morioka M, Murai Y, Tanaka E. Clinical neuroprotective drugs for treatment and prevention of stroke. Int J Mol Sci. (2012) 13:7739–61. 10.3390/ijms13067739
    1. Hess DC, Fagan SC. Repurposing an old drug to improve the use and safety of tissue plasminogen activator for acute ischemic stroke: Minocycline. Pharmacotherapy (2010) 30:55s−61s. 10.1592/phco.30.pt2.55S
    1. Zhao F, Xi G, Liu W, Keep RF, Hua Y. Minocycline attenuates iron-induced brain injury. Acta Neuroch. (2016) 121:361–5. 10.1007/978-3-319-18497-5_62
    1. Fagan SC, Waller JL, Nichols FT, Edwards DJ, Pettigrew LC, Clark WM, et al. . Minocycline to improve neurologic outcome in stroke (minos): a dose-finding study. Stroke (2010) 41:2283–7. 10.1161/STROKEAHA.110.582601
    1. Goldstein LB, Bertels C, Davis JN. Interrater reliability of the nih stroke scale. Arch Neurol. (1989) 46:660–2. 10.1001/archneur.1989.00520420080026
    1. Balu S. Differences in psychometric properties, cut-off scores, and outcomes between the barthel index and modified rankin scale in pharmacotherapy-based stroke trials: systematic literature review. Curr Med Res Opin. (2009) 25:1329–41. 10.1185/03007990902875877
    1. Wilkinson PR, Wolfe CD, Warburton FG, Rudd AG, Howard RS, Ross-Russell RW, et al. Longer term quality of life and outcome in stroke patients: is the barthel index alone an adequate measure of outcome? Quality Health Care (1997) 6:125–130.
    1. Kwon S, Hartzema AG, Duncan PW, Min-Lai S. Disability measures in stroke: Relationship among the barthel index, the functional independence measure, and the modified rankin scale. Stroke (2004) 35:918–23. 10.1161/01.STR.0000119385.56094.32
    1. Kwah LK, Diong J. National institutes of health stroke scale (nihss). J Physiother. (2014) 60:61. 10.1016/j.jphys.2013.12.012
    1. Alba AC, Alexander PE, Chang J, MacIsaac J, DeFry S, Guyatt GH. High statistical heterogeneity is more frequent in meta-analysis of continuous than binary outcomes. J Clin Epidemiol. (2016) 70:129–35. 10.1016/j.jclinepi.2015.09.005
    1. von Hippel PT. The heterogeneity statistic i(2) can be biased in small meta-analyses. BMC Med Res Methodol. (2015) 15:35 10.1186/s12874-015-0024-z
    1. Olavarria VV, Delgado I, Hoppe A, Brunser A, Carcamo D, Diaz-Tapia V, et al. . Validity of the nihss in predicting arterial occlusion in cerebral infarction is time-dependent. Neurology (2011) 76:62–8. 10.1212/WNL.0b013e318203e977
    1. Hannan EL. Randomized clinical trials and observational studies: Guidelines for assessing respective strengths and limitations. JACC (2008) 1:211–7. 10.1016/j.jcin.2008.01.008
    1. Shrier I, Boivin JF, Steele RJ, Platt RW, Furlan A, Kakuma R, et al. . Should meta-analyses of interventions include observational studies in addition to randomized controlled trials? A critical examination of underlying principles. Am J Epidemiol. (2007) 166:1203–9. 10.1093/aje/kwm189
    1. Walter SD. Variation in baseline risk as an explanation of heterogeneity in meta-analysis. Statist Med. (1997) 16:2883–900. 10.1002/(SICI)1097-0258(19971230)16:24<2883::AID-SIM825>;2-B
    1. Burchell SR, Dixon BJ, Tang J, Zhang JH. Isoflurane provides neuroprotection in neonatal hypoxic ischemic brain injury. J Invest Med. (2013) 61:1078–83. 10.2310/JIM.0b013e3182a07921

Source: PubMed

3
Tilaa