Pharmacological brain cytoprotection in acute ischaemic stroke - renewed hope in the reperfusion era

Marc Fisher, Sean I Savitz, Marc Fisher, Sean I Savitz

Abstract

For over 40 years, attempts to develop treatments that protect neurons and other brain cells against the cellular and biochemical consequences of cerebral ischaemia in acute ischaemic stroke (AIS) have been unsuccessful. However, the advent of intravenous thrombolysis and endovascular thrombectomy has taken us into a new era of treatment for AIS in which highly effective reperfusion therapy is widely available. In this context, cytoprotective treatments should be revisited as adjunctive treatment to reperfusion therapy. Renewed efforts should focus on developing new drugs that target multiple aspects of the ischaemic cascade, and previously developed drugs should be reconsidered if they produced robust cytoprotective effects in preclinical models and their safety profiles were reasonable in previous clinical trials. Several development pathways for cytoprotection as an adjunct to reperfusion can be envisioned. In this Review, we outline the targets for cytoprotective therapy and discuss considerations for future drug development, highlighting the recent ESCAPE-NA1 trial of nerinetide, which produced the most promising results to date. We review new types of clinical trial to evaluate whether cytoprotective drugs can slow infarct growth prior to reperfusion and/or ameliorate the consequences of reperfusion, such as haemorrhagic transformation. We also highlight how advanced brain imaging can help to identify patients with salvageable ischaemic tissue who are likely to benefit from cytoprotective therapy.

Conflict of interest statement

M.F. serves on the data, safety monitoring board for the ESCAPE-Next trial. He also serves as a consultant to Simcere USA and Lumosa of Taiwan. As an employee of the institution (UTHealth), S.I.S. has served in the following roles: as a site investigator in clinical trials sponsored by industry companies (Athersys, ReNeuron, San Bio, KM Pharma, Abbvie) for which UTHealth receives payments on the basis of clinical trial contracts; as an investigator on clinical trials supported by NIH grants, the Department of Defense, Let’s Cure CP, the TIRR Foundation, and the Cord Blood Registry Systems; as a principal investigator or co-investigator on NIH-funded grants in basic science and clinical research; and as principal investigator for an imaging analysis center for clinical trials sponsored by SanBio and ReNeuron. In his capacity as a UTHealth employee, Dr. Savitz has provided consulting services on behalf of UTHealth to ReNeuron, Lumosa, Deck Therapeutics, KM Pharma, Neurexcell, Abbvie, TMC Biodesign and ArunA. All compensation from such consulting arrangements have been paid to UTHealth.

© 2022. Springer Nature Limited.

Figures

Fig. 1. Elements of the ischaemic cascade…
Fig. 1. Elements of the ischaemic cascade in the neurovascular unit during acute ischaemic stroke.
Only selected elements are shown, and the signalling evolves over time after the onset of ischaemia. Oxygen and glucose deprivation leads to release of glutamate from astrocytes (1), which activates excitotoxic signalling within neurons, leading to neuronal injury and death, and activates microglia. Neuronal injury leads to the release of damage-associated molecular patterns (DAMPs), which also activate microglia (2), which consequently release pro-inflammatory cytokines that lead to further damage of neurons and promote endothelial activation and blood–brain barrier (BBB) opening. DAMPS also directly promote BBB breakdown. Leukocyte trafficking and migration to the ischaemic region (3) lead to further release of cytokines, amplification of neuroinflammation and continued neuronal damage. In parallel, neurons release ‘help me’ signals (4) that activate astrocytes and microglia to release factors that protect neurons. MMPs, matrix metalloproteinases; TH1 cell, T helper 1 cell; TH17 cell, T helper 17 cell.
Fig. 2. Outcomes of acute ischaemic stroke…
Fig. 2. Outcomes of acute ischaemic stroke with and without cytoprotective therapy and reperfusion.
The aim of cytoprotective therapy administered as early as possible after stroke onset, is to ‘freeze’ the penumbra (left) so that the infarct core does not grow further before definitive reperfusion therapy can be completed. This protection minimizes the size of the final infarct (bottom left). Without cytoprotective therapy, the final infarct size could be larger despite successful reperfusion (bottom right). Cytoprotective therapy can only be beneficial if reperfusion is achieved before the protective effects diminish. If reperfusion therapy is not administered within this time, the final infarct could be as large as it would have been without cytoprotection (bottom centre). Reperfusion injury can lead to haemorrhagic transformation regardless of whether cytoprotective therapy is used.

References

    1. Moskowitz MA, Lo EH, Iadecola C. The science of stroke: mechanisms in search of treatments. Neuron. 2010;67:181–198.
    1. Savitz SI, Baron JC, Fisher M. Stroke Treatment Academic Industry Roundtable X: brain cytoprotection therapies in the reperfusion era. Stroke. 2019;50:1026–1031.
    1. Lyden P, Buchan A, Boltze J, Fisher M. Top priorities for cerebroprotective studies–a paradigm shift: report from STAIR XI. Stroke. 2021;52:3063–3071.
    1. Savitz SI, Fisher M. Future of neuroprotection for acute stroke: in the aftermath of the SAINT trials. Ann. Neurol. 2007;61:396–402.
    1. Tymianski M. Combining neuroprotection with endovascular treatment of acute stroke: is there hope? Stroke. 2017;48:1700–1705.
    1. Hill MD, et al. Efficacy and safety of nerinetide for the treatment of acute ischaemic stroke (ESCAPE-NA1): a multicentre, double-blind, randomised controlled trial. Lancet. 2020;395:878–887.
    1. Xing C, Arai K, Lo EH, Hommel M. Pathophysiologic cascades in ischemic stroke. Int. J. Stroke. 2012;7:378–385.
    1. Griffin JH, Fernandez JA, Lyden PD, Zlokovic BV. Activated protein C promotes neuroprotection: mechanisms and translation to the clinic. Thromb. Res. 2016;141:S62–S64.
    1. Cui H, et al. PDZ protein interactions underlying NMDA receptor-mediated excitotoxicity and neuroprotection by PSD-95 inhibitors. J. Neurosci. 2007;2:9901–9915.
    1. Lyden P, et al. Results of the ICTuS 2 trial (Intravascular Cooling in the Treatment of Stroke 2) Stroke. 2016;47:2888–2895.
    1. Bai J, Lyden PD. Revisiting cerebral postischemic reperfusion injury: new insights in understanding reperfusion failure, hemorrhage, and edema. Int. J. Stroke. 2015;10:143–152.
    1. Jung JE, et al. Reperfusion and neurovascular dysfunction in stroke: from basic mechanisms to potential strategies for neuroprotection. Mol. Neurobiol. 2010;41:172–179.
    1. Wang Q, Tang XN, Yenari MA. The inflammatory response in stroke. J. Neuroimmunol. 2007;184:53–68.
    1. Bernardo-Castro S, et al. Pathophysiology of blood–brain barrier permeability throughout the different stages of ischemic stroke and its implication on hemorrhagic transformation and recovery. Front. Neurol. 2020;11:594672.
    1. Yang Y, Rosenberg GA. Matrix metalloproteinases as therapeutic targets for stroke. Brain Res. 2015;1623:30–38.
    1. Molina CA, et al. Timing of spontaneous recanalization and risk of hemorrhagic transformation in acute cardioembolic stroke. Stroke. 2001;32:1079–1084.
    1. Shuaib A, Butcher K, Mohammad AA, Saqqur M, Liebeskind DS. Collateral blood vessels in acute ischaemic stroke: a potential therapeutic target. Lancet Neurol. 2011;10:909–921.
    1. Bornstein NM, et al. An injectable implant to stimulate the sphenopalatine ganglion for treatment of acute ischaemic stroke up to 24h from onset (ImpACT-24B): an international, randomised, double-blind, sham-controlled, pivotal trial. Lancet. 2019;394:219–229.
    1. Elkind MSV, et al. Natalizumab in acute ischemic stroke (ACTION II): a randomized, placebo-controlled trial. Neurology. 2020;95:e1091–e1104.
    1. Poungvarin N, et al. Effects of dexamethasone in primary supratentorial intracerebral hemorrhage. N. Engl. J. Med. 1987;316:1229–1233.
    1. Mays RW, Savitz SI. Intravenous cellular therapies for acute ischemic stroke. Stroke. 2018;49:1058–1065.
    1. Henninger N, Kumar R, Fisher M. Acute ischemic stroke therapy. Expert. Rev. Cardiovasc. Ther. 2010;8:1389–1398.
    1. Bosetti F, et al. Translational stroke research: vision and opportunities. Stroke. 2017;48:2632–2637.
    1. Fisher M, et al. Update of the stroke therapy academic industry roundtable preclinical recommendations. Stroke. 2009;40:2244–2250.
    1. Dirnagl U, et al. A concerted appeal for international cooperation in preclinical stroke research. Stroke. 2013;44:1754–1760.
    1. Hill MD, et al. Safety and efficacy of NA-1 in patients with iatrogenic stroke after endovascular aneurysm repair (ENACT): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2012;11:942–950.
    1. Mayor-Nunez D, et al. Plasmin-resistant PSD-95 inhibitors resolve effect-modifying drug–drug interactions between alteplase and nerinetide in acute stroke. Sci. Transl. Med. 2021;13(588):eabb1498.
    1. US National Library of Medicine. (2020).
    1. Ehrenreich H, et al. Recombinant human erythropoietin in the treatment of acute ischemic stroke. Stroke. 2009;40:e647–e656.
    1. Worthmann H, et al. Asymmetric dimethylarginine in response to recombinant tissue-type plasminogen activator and erythropoietin in acute stroke. Stroke. 2013;44:2128–2133.
    1. Jia L, Chopp M, Zhang L, Lu M, Zhang Z. Erythropoietin in combination of tissue plasminogen activator exacerbates brain hemorrhage when treatment is initiated 6h after stroke. Stroke. 2010;41:2071–2076.
    1. Riou-Comte N, et al. Direct transfer to angiosuite for patients with severe acute stroke treated with thrombectomy: the multicentre randomised controlled DIRECT ANGIO trial protocol. BMJ Open. 2021;11:e040522.
    1. Link TW, Santillan A, Patsalides A. Intra-arterial neuroprotective therapy as an adjunct to endovascular intervention in acute ischemic stroke: a review of the literature and future directions. Interv. Neuroradiol. 2020;26:405–415.
    1. Saver JL, et al. Prehospital use of magnesium sulfate as neuroprotection in acute stroke. N. Engl. J. Med. 2015;372:528–536.
    1. Vidale S, Agostoni E. Prehospital stroke scales and large vessel occlusion: a systematic review. Acta Neurol. Scand. 2018;138:24–31.
    1. Smith EE, et al. Accuracy of prediction instruments for diagnosing large vessel occlusion in individuals with suspected stroke: a systematic review for the 2018 guidelines for the early management of patients with acute ischemic stroke. Stroke. 2018;49:e111–e122.
    1. Goyal N, et al. FABS: an intuitive tool for screening of stroke mimics in the emergency department. Stroke. 2016;47:2216–2220.
    1. Sergot PB, et al. Portable neuromonitoring device detects large vessel occlusion in suspected acute ischemic stroke. Stroke. 2021;52:1437–1440.
    1. Grotta JC, et al. Prospective, multicenter, controlled trial of mobile stroke units. N. Engl. J. Med. 2021;385:971–981.
    1. Ebinger M, et al. Association between dispatch of mobile stroke units and functional outcomes among patients with acute ischemic stroke in Berlin. JAMA. 2021;325:454–466.
    1. Savitz SI, Baron JC, Yenari MA, Sanossian N, Fisher M. Reconsidering neuroprotection in the reperfusion era. Stroke. 2017;48:3413–3419.
    1. Wu TC, et al. IAT-TiMeS: intra-arterial thrombectomy transfer metric study in Texas. J. Stroke Cerebrovasc. Dis. 2021;30:105602.
    1. Wu TC, et al. Telemedicine-guided remote enrollment of patients into an acute stroke trial. Ann. Clin. Transl. Neurol. 2015;2:38–42.
    1. Reddy ST, et al. Rapid infarct progression in anterior circulation large vessel occlusion ischemic stroke patients during inter-facility transfer. J. Stroke Cerebrovasc. Dis. 2020;29:105308.
    1. Boulouis G, et al. Clinical imaging factors associated with infarct progression in patients with ischemic stroke during transfer for mechanical thrombectomy. JAMA Neurol. 2017;74:1361–1367.
    1. Ospel JM, et al. Which acute ischemic stroke patients are fast progressors? Results from the ESCAPE trial control arm. Stroke. 2021;52:1847–1850.
    1. Reddy ST, et al. Patients transferred within a telestroke network for large-vessel occlusion. J. Telemed. Telecare. 2020 doi: 10.1177/1357633X20957894.
    1. Liebeskind DS. Neuroprotection from a collateral perspective. IDrugs. 2005;8:222–228.
    1. Albers GW, et al. Thrombectomy for stroke at 6 to 16 hours with selection by perfusion imaging. N. Engl. J. Med. 2018;378:708–718.
    1. Nogueira RG, et al. Thrombectomy 6 to 24 hours after stroke with a mismatch between deficit and infarct. N. Engl. J. Med. 2018;378:11–21.
    1. Kim Y, et al. Utilization and availability of advanced imaging in patients with acute ischemic stroke. Circ. Cardiovasc. Qual. Outcomes. 2021;14:e006989.
    1. Sheth SA, et al. Machine learning-enabled automated determination of acute ischemic core from computed tomography angiography. Stroke. 2019;50:3093–3100.
    1. Cahn L, et al. Deployment of portable, bedside, low-field magnetic resonance imaging for evaluation of stroke patients [abstract] Neurology. 2020;94(15 Suppl.):272.
    1. Sheth KN, et al. Assessment of brain injury using portable, low-field magnetic resonance imaging at the bedside of critically ill patients. JAMA Neurol. 2021;78:41–47.
    1. Lu G, He Q, Shen Y, Cao F. Potential biomarkers for predicting hemorrhagic transformation of ischemic stroke. Int. J. Neurosci. 2018;128:79–89.
    1. Luby M, et al. Frequency of blood–brain barrier disruption post-endovascular therapy and multiple thrombectomy passes in acute ischemic stroke patients. Stroke. 2019;50:2241–2244.
    1. Stroke Preclinical Assessment Network. Interventions (2021).

Source: PubMed

3
구독하다