Rationale and methods of the Antioxidant and NMDA receptor blocker Weans Anoxic brain damage of KorEa OHCA patients (AWAKE) trial

Jin-Ho Choi, Byeong Jo Chun, Seok Ran Yeom, Sung Phil Chung, Young Hwan Lee, Yun-Hee Kim, Ji Sung Lee, Jin Hwan Lee, Hwan Goo Lee, Jing Yu Jin, Chun San An, Byoung Joo Gwag, Jin-Ho Choi, Byeong Jo Chun, Seok Ran Yeom, Sung Phil Chung, Young Hwan Lee, Yun-Hee Kim, Ji Sung Lee, Jin Hwan Lee, Hwan Goo Lee, Jing Yu Jin, Chun San An, Byoung Joo Gwag

Abstract

Background: Ischemic brain injury is a major hurdle that limits the survival of resuscitated out-of-hospital cardiac arrest (OHCA).

Methods: The aim of this study is to assess the feasibility and potential for reduction of ischemic brain injury in adult OHCA patients treated with high- or low-dose Neu2000K, a selective blocker of N-methyl-D-aspartate (NMDA) type 2B receptor and also a free radical scavenger, or given placebo. This study is a phase II, multicenter, randomized, double-blinded, prospective, intention-to-treat, placebo-controlled, three-armed, safety and efficacy clinical trial. This trial is a sponsor-initiated trial supported by GNT Pharma. Successfully resuscitated OHCA patients aged 19 to 80 years would be included. The primary outcome is blood neuron-specific enolase (NSE) level on the 3rd day. The secondary outcomes are safety, efficacy defined by study drug administration within 4 h in > 90% of participants, daily NSE up to 5th day, blood S100beta, brain MRI apparent diffusion coefficient imaging, cerebral performance category (CPC), and Modified Rankin Scale (mRS) at 5th, 14th, and 90th days. Assuming NSE of 42 ± 80 and 80 ± 80 μg/L in the treatment (high- and low-dose Neu2000K) and control arms with 80% power, a type 1 error rate of 5%, and a 28% of withdrawal prior to the endpoint, the required sample size is 150 patients.

Discussion: The AWAKE trial explores a new multi-target neuroprotectant for the treatment of resuscitated OHCA patients.

Trial registration: ClinicalTrials.gov NCT03651557 . Registered on August 29, 2018.

Keywords: Free radical scavenger; Ischemic-reperfusion brain injury; NMDA antagonist; Neu2000K; Out-of-hospital cardiac arrest; Randomized controlled trial.

Conflict of interest statement

J.L., G.L., J.J., C.A., and B.G. are employees of GNT Pharma. All the other authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Mechanism of Neu2000K as a dual-target neuroprotectant for ischemic and reperfusion brain injury. In patients suffering from cardiac arrest, oxygen and glucose are deprived from the brain, which results in glutamate release and accumulation at the synaptic cleft. Excess glutamate causes acute and fulminant neuronal death through the overactivation of calcium ion-permeable NMDA receptors. Free radicals such as superoxide and hydrogen peroxide are produced in the mitochondria over hours and days after the return of spontaneous circulation. Such prolonged and excessive oxidative stress causes delayed cell death including the neurons and glia. Neu2000K moderately and selectively blocks the NR2B subtype of NMDA receptor, which activates pro-survival signaling pathways without causing excessive calcium influx, which can reduce acute neuronal death. Neu2000K also functions as a potent reactive oxygen species scavenger and reduces oxidative stress or reperfusion injury, which can reduce delayed brain cell death
Fig. 2
Fig. 2
Study flow. OHCA, out-of-hospital cardiac arrest; ROSC, return of spontaneous circulation; ECMO, extracorporeal membranous oxygenation; TTM, targeted temperature management; CT, computed tomography; NSE, neuron-specific enolase; MRI, magnetic resonance imaging; CPC, cerebral performance category; mRS, Modified Rankin Scale

References

    1. Berdowski J, Berg RA, Tijssen JGP, et al. Global incidences of out-of-hospital cardiac arrest and survival rates: systematic review of 67 prospective studies. Resuscitation. 2010;81:1479–1487. doi: 10.1016/j.resuscitation.2010.08.006.
    1. Virani SS, Alonso A, Aparicio HJ, et al. Heart disease and stroke statistics—2021 update. Circulation. 2021;143:e254–e743. doi: 10.1161/CIR.0000000000000950.
    1. Kim JY, Hwang SOSS, Shin S Do, et al. Korean Cardiac Arrest Research Consortium (KoCARC): rationale, development, and implementation. Clin Exp Emerg Med. 2018;5:165–176. doi: 10.15441/ceem.17.259.
    1. Gräsner J-T, Wnent J, Herlitz J, et al. Survival after out-of-hospital cardiac arrest in Europe - results of the EuReCa TWO study. Resuscitation. 2020;148:218–226. doi: 10.1016/j.resuscitation.2019.12.042.
    1. Dennis M, Lal S, Forrest P, et al. In-depth extracorporeal cardiopulmonary resuscitation in adult out-of-hospital cardiac arrest. J Am Heart Assoc. 2020;9:e016521. doi: 10.1161/JAHA.120.016521.
    1. Kilgannon JH, Roberts BW, Jones AE, et al. Arterial blood pressure and neurologic outcome after resuscitation from cardiac arrest*. Crit Care Med. 2014;42:2083–2091. doi: 10.1097/CCM.0000000000000406.
    1. Choi DW. Excitotoxicity: still hammering the ischemic brain in 2020. Front Neurosci. 2020;14:1104. doi: 10.3389/fnins.2020.579953.
    1. Vieira M, Yong XLH, Roche KW, et al. Regulation of NMDA glutamate receptor functions by the GluN2 subunits. J Neurochem. 2020;154:121–143. doi: 10.1111/jnc.14970.
    1. Stanika RI, Pivovarova NB, Brantner CA, et al. Coupling diverse routes of calcium entry to mitochondrial dysfunction and glutamate excitotoxicity. Proc Natl Acad Sci. 2009;106:9854–9859. doi: 10.1073/pnas.0903546106.
    1. Pose-Utrilla J, García-Guerra L, Del Puerto A, et al. Excitotoxic inactivation of constitutive oxidative stress detoxification pathway in neurons can be rescued by PKD1. Nat Commun. 2017;8:2275. doi: 10.1038/s41467-017-02322-5.
    1. Cho SJ, Park UJ, Chung J-M, et al. Neu 2000, an NR2B-selective, moderate NMDA receptor antagonist and potent spin trapping molecule for stroke. Drug News Perspect. 2010;23:549. doi: 10.1358/dnp.2010.23.9.1513493.
    1. Wu QJ, Tymianski M. Targeting NMDA receptors in stroke: new hope in neuroprotection. Mol Brain. 2018;11:15. doi: 10.1186/s13041-018-0357-8.
    1. Lai TW, Zhang S, Wang YT. Excitotoxicity and stroke: identifying novel targets for neuroprotection. Prog Neurobiol. 2014;115:157–188. doi: 10.1016/j.pneurobio.2013.11.006.
    1. Ginsberg MD. Neuroprotection for ischemic stroke: past, present and future. Neuropharmacology. 2008;55:363–389. doi: 10.1016/j.neuropharm.2007.12.007.
    1. Yoon WJ, Won SJ, Ryu BR, et al. Blockade of ionotropic glutamate receptors produces neuronal apoptosis through the Bax-cytochrome C-caspase pathway: the causative role of Ca2+ deficiency. J Neurochem. 2003;85:525–533. doi: 10.1046/j.1471-4159.2003.01724.x.
    1. Gwag BJ, Lobner D, Koh JY, et al. Blockade of glutamate receptors unmasks neuronal apoptosis after oxygen-glucose deprivation in vitro. Neuroscience. 1995;68:615–619. doi: 10.1016/0306-4522(95)00232-8.
    1. Seo SY, Kim EY, Kim H, et al. Neuroprotective effect of high glucose against NMDA, free radical, and oxygen–glucose deprivation through enhanced mitochondrial potentials. J Neurosci. 1999;19:8849. doi: 10.1523/JNEUROSCI.19-20-08849.1999.
    1. Shin JH, Cho SI, Lim HR, et al. Concurrent administration of Neu 2000 and lithium produces marked improvement of motor neuron survival, motor function, and mortality in a mouse model of amyotrophic lateral sclerosis. Mol Pharmacol. 2007;71:965–975. doi: 10.1124/mol.106.030676.
    1. Lee J-M, Zipfel GJ, Choi DW. The changing landscape of ischaemic brain injury mechanisms. Nature. 1999;399:A7–A14. doi: 10.1038/399a007.
    1. Dyker AG, Edwards KR, Fayad PB, et al. Safety and tolerability study of aptiganel hydrochloride in patients with an acute ischemic stroke. Stroke. 1999;30:2038–2042. doi: 10.1161/01.STR.30.10.2038.
    1. Diener H-C, AlKhedr A, Busse O, et al. Treatment of acute ischaemic stroke with the low-affinity, use-dependent NMDA antagonist AR-R15896AR. J Neurol. 2002;249:561–568. doi: 10.1007/s004150200065.
    1. Gwag BJ, Lee YA, Ko SY, et al. Marked prevention of ischemic brain injury by Neu 2000, an NMDA antagonist and antioxidant derived from aspirin and sulfasalazine. J Cereb Blood Flow Metab. 2007;27:1142–1151. doi: 10.1038/sj.jcbfm.9600418.
    1. Kvrivishvili G. Glycine and neuroprotective effect of hypothermia in hypoxic–ischemic brain damage. NeuroReport. 2002;13:1995–2000. doi: 10.1097/00001756-200211150-00001.
    1. Ooboshi H, Ibayashi S, Takano K, et al. Hypothermia inhibits ischemia-induced efflux of amino acids and neuronal damage in the hippocampus of aged rats. Brain Res. 2000;884:23–30. doi: 10.1016/S0006-8993(00)02861-4.
    1. González-Ibarra FP, Varon J, López-Meza EG. Therapeutic hypothermia: critical review of the molecular mechanisms of action. Front Neurol. 2011;2:4. doi: 10.3389/fneur.2011.00004.
    1. Hardingham GE. Coupling of the NMDA receptor to neuroprotective and neurodestructive events. Biochem Soc Trans. 2009;37:1147–1160. doi: 10.1042/BST0371147.
    1. Papadia S, Soriano FX, Léveillé F, et al. Synaptic NMDA receptor activity boosts intrinsic antioxidant defenses. Nat Neurosci. 2008;11:476–487. doi: 10.1038/nn2071.
    1. Visavadiya NP, McEwen ML, Pandya JD, et al. Antioxidant properties of Neu 2000 on mitochondrial free radicals and oxidative damage. Toxicol Vitr. 2013;27:788–797. doi: 10.1016/j.tiv.2012.12.011.
    1. Park UJ, Lee YA, Won SM, et al. Blood-derived iron mediates free radical production and neuronal death in the hippocampal CA1 area following transient forebrain ischemia in rat. Acta Neuropathol. 2011;121:459–73. doi: 10.1007/s00401-010-0785-8.
    1. Won SM, Lee JH, Park UJ, et al. Iron mediates endothelial cell damage and blood-brain barrier opening in the hippocampus after transient forebrain ischemia in rats. Exp Mol Med. 2011;43:121. doi: 10.3858/emm.2011.43.2.020.
    1. Eastwood GM, Schneider AG, Suzuki S, et al. Targeted therapeutic mild hypercapnia after cardiac arrest: A phase II multi-centre randomised controlled trial (the CCC trial) Resuscitation. 2016;104:83–90. doi: 10.1016/j.resuscitation.2016.03.023.
    1. Meyer MAS, Wiberg S, Grand J, et al. Treatment effects of interleukin-6 receptor antibodies for modulating the systemic inflammatory response after out-of-hospital cardiac arrest (The IMICA trial): A double-blinded, placebo-controlled, single-center, randomized, clinical trial. Circulation. 2021;143:1841–1851. doi: 10.1161/CIRCULATIONAHA.120.053318.

Source: PubMed

3
Prenumerera