Skip to main content

Abstract

The treatment of acute ischemic stroke continues to advance. The mainstay of treatment remains intravenous thrombolysis with alteplase. Recent studies demonstrated that later treatment with alteplase is beneficial in patients selected with advanced imaging techniques. Tenecteplase has been evaluated as an alternative thrombolytic drug and evidence suggests that it is as least as effective as alteplase and may lyse large vessel clots more effectively. Endovascular therapy with mechanical thrombectomy has now been shown to be beneficial up to 24 hours after stroke onset in carefully selected patients with proximal, large vessel occlusions. Ongoing studies are evaluating the effectiveness of thrombectomy in patients with more distal vessel occlusions and patients with proximal large vessel occlusions with larger ischemic core volumes and also in patients with milder neurological deficits. Cytoprotection is another potential acute stroke therapy that has not demonstrated efficacy in prior clinical trials. It should be reconsidered as an adjunct to reperfusion and a variety of new clinical trials can be envisioned to evaluate the potential benefits of cytoprotection in patients before and after reperfusion.
Over the past 5 years since the publication of the prior review on this topic in Circulation Research, progress continues to be made in the field of acute ischemic stroke therapy.1,2 Therefore, the focus of this update will be on these advances and what future advances may occur. Fundamentally, the goal of acute ischemic stroke therapy is to salvage as much of the ischemic region at risk of infarction as possible to save brain and improve functional outcome.3 The acute occlusion of an extracranial or intracranial blood vessel by an in-situ thrombus or embolus from the heart or a proximal large blood vessel initiates a complex cascade event, that is, the ischemic cascade, as illustrated by Figure 1 that ultimately leads to infarction in a variable portion of the ischemic tissue. Ischemia affects not only neurons but also astrocytes, the microglia, pericytes, and the endothelial lining of blood vessels in the ischemic region.4 This conglomeration of cells affected by ischemia is termed the neurovascular unit and will be discussed as all of them are potential targets for therapies, that is, cytoprotection. The evolution of ischemic injury is quite variable among individual ischemic stroke patients and the pace of progression to irreversible ischemic injury, that is, infarction is dependent upon factors such as the adequacy of the collateral circulation, blood pressure, the metabolic milieu, temperature, and the age of the patient.5 With the help of advanced imaging techniques such as computed tomography (CT) perfusion and magnetic resonance imaging (MRI) diffusion/perfusion imaging this evolution can now be observed in individual patients.6 The region of irreversible injury or ischemic core and ischemic tissue at risk of infarction, the ischemic penumbra, can be approximated on both imaging techniques and this type of imaging provides clinicians with important information about the status of the ischemic brain and for potentially making treatment decisions, as will be discussed. The severity of ischemia in different portions of the ischemic territory can also be identified, and this information predicts the rapidity of progression to infarction in individual patients.7 Progression of ischemic injury is quite variable among ischemic stroke patients and this variability is termed rapid, intermediate, and slow progression. Approaches to treatment based upon these patterns of ischemic evolution will likely vary and future clinical trials will account for them in trial design and choice of therapeutic agents to be evaluated.
Figure 1. A schematic depiction of the ischemic cascade (courtesy of Dr Sean Savitz and Dr Nikunj Satani, UT-Health Houston). DAMP indicates deoxyadenosine-5-monophosphate; IFN, interferon; IL, interleukin; MMP indicates matrix metalloproteinase; Th, helper T cell; and TNF, tumor necrosis factor. Illustration credit: Ben Smith.
Currently, 2 approved therapies for acute ischemic stroke are available and both are intended to reperfuse the ischemic region. The first therapy to be developed was intravenous thrombolysis with tPA (tissue-type plasminogen activator) also known as alteplase.8 The second approach is a minimally invasive procedure, ie, mechanical thrombectomy. This endovascular therapy (EVT) using stent-retrievers has demonstrated efficacy in opening occluded proximal blood vessels and improving functional outcome in patients with large vessel occlusions (LVO) to remove the clot (Figure 2).9 A third approach to acute ischemic stroke therapy has been attempting to impede the ischemic cascade, termed neuroprotection in the past.10 All efforts to develop such treatments were unsuccessful in many clinical trials. New approaches to neuroprotection should now be termed cytoprotection as they should target components of the neurovascular unit beyond neurons.11 Therapies with multiple mechanisms of action will be favored in future clinical trials because of the complexity of the ischemic cascade and the multiplicity of the cellular targets. In this review, we will discuss all three of these approaches to acute ischemic stroke therapy and how they may be used synergistically.
Figure 2. Thrombectomy for acute ishemic stroke due to large vessel occlusion. A, Computed tomography (CT) angiographic study shows an occlusion of the right middle cerebral artery (MCA) M1 segment (arrow). B–D, CT shows a hyperdense M1 associated with a thrombus (arrow). No ischemic core visible, Alberta Stroke Program Early CT Score 10. E, CT perfusion scan shows no core infarct (cerebral blood flow [CBF] <30%) and a large penumbra (arrow) with a mismatch volume of 34 mL. F–G, Right internal carotid artery angiogram shows an occluded right MCA (arrow) with a Treatment in Cerebral Ischemia Score (TICI)=0 flow and following thrombectomy a complete revascularization with a TICI=3 flow. H, Follow-up diffusion-weighted–magnetic resonance imaging shows a small ischemic area in right globus pallidus (arrow).

Intravenous Thrombolysis

Intravenous thrombolysis (IVT) is the mainstay of acute ischemic stroke therapy. Over the past 5 years, IVT has been moving from a purely time window to include a tissue clock approach, the development of new thrombolytic drugs and to determine whether IVT should be skipped or used as bridging therapy before thrombectomy for patients with anterior circulation LVO.

Time Is Brain

The phrase time is brain emphasizes that human nervous system tissue is rapidly lost as stroke progresses with 1.9 million neurons lost per minute in LVO and emergent evaluation and therapy are required.12 Over time strategies have been developed to reduce the time delay from symptom onset to treatment. Stroke recognition scales used in the prehospital setting improve the recognition and diagnosis of stroke, thereby aiding emergency service triage of stroke patients. Face Arm Speech Time and the Melbourne Ambulance Stroke Screen were found to be the most sensitive for stroke recognition, and the CPSS (Cincinnati Prehospital Stroke Scale) had higher specificity.13 In the emergency medical services (EMS) setting, the Los Angeles Motor Scale and Rapid Arterial Occlusion Evaluation were the best scoring scales with the highest accuracy for stroke due to LVO.14
The optimal transfer paradigm for emergent LVO strokes is still controversial. Two transfer models have been developed: (1) transport the patient directly to the nearest comprehensive stroke center to receive tPA and, if appropriate, immediate EVT (mothership model) or (2) transport the patient to the nearest primary stroke center to receive tPA and then transfer appropriate patients to the nearest comprehensive stroke center for EVT (drip and ship model). The RACECAT trial (Direct Transfer to Endovascular Center of Acute Stroke Patients With Suspected Large Vessel Occlusion in the Catalan Territory; https://www.clinicaltrials.gov; Unique identifier: NCT02795962) found that the modified Rankin Scale (mRS) score at 90 days was comparable with these two approaches (adjusted odds ratio [OR], 1.02 [95% CI, 0.8–1.2]).15 The Mission: Lifeline Severity–based Stroke Triage Algorithm for EMS recommended direct transport to a comprehensive stroke center if the travel time from pick up point to the comprehensive stroke center was <30 minutes. In rural communities or those where large distances separate stroke centers causing additional transport time, medical transport should be considered.16 Other than transporting patients by EMS, a telestroke service has become more and more important for rural stroke centers and primary stroke centers that cannot perform EVT and advanced imaging. Telestroke was superior to telephone-based consultation for increasing the rate of correct thrombolysis eligibility decisions and rate of intravenous tPA usage.17
New technologies are emerging. Mobile stroke units (MSU) have computed tomographic scanning with or without angiography, point-of-care laboratory testing, and thrombolysis capabilities on board. The B_PROUD study (Berlin_Prehospital or Usual Delivery in Stroke Care) was a prospective, nonrandomized, controlled intervention study in Germany and found that the dispatch of an MSU, compared with conventional ambulances alone, was associated with less disability at 3 months (common OR for worse mRS, 0.71 [95% CI, 0.58–0.86]) in 1543 acute ischemic stroke eligible for the indication of IVT or EVT.18 In the United States, the BEST MSU study (The Benefits of Stroke Treatment Delivered by a Mobile Stroke Unit Compared with Standard Management by Emergency Medical Services) was an observational, prospective, multicenter, alternating-week trial that enrolled 1515 patients of whom 1047 were eligible to receive tPA; 617 received care in the MSU; and 430 by standard EMS. The utility-weighted disability outcomes at 90 days were better with MSU care than with EMS (adjusted OR, 2.43 [95% CI, 1.75–3.36]) in patients with acute stroke who were eligible for tPA.19 These 2 important studies of MSU utility demonstrated that MSU care shortened the time from onset to thrombolysis, increased the rate of thrombolysis, optimized prehospital triage, and improved patients’ 3-month clinical outcomes. MSU care may also be used for the emergency treatment of other cerebrovascular diseases in the future.

Time Window Versus a Tissue Clock Approach

The WAKE-UP trial (Efficacy and Safety of MRI-Based Thrombolysis in Wake-Up Stroke) demonstrated that alteplase is beneficial for wake-up or unknown onset time patients with an acute ischemic lesion on diffusion-weighted imaging (DWI) but no parenchymal hyperintensity with standard window settings on a fluid-attenuated inversion recovery (FLAIR) MRI sequence. Treatment had to occur within 4.5 hours of symptom onset when compared with placebo (mRS 0–1, and an adjusted OR, 1.61 [95% CI, 1.09–2.36]) was observed.20 It did not significantly increase the risk of symptomatic intracerebral hemorrhage (sICH) or death.20 The DWI-FLAIR mismatch was presumed to identify a stroke onset time within 4.5 hours, had a 62% (95% CI, 57–67) sensitivity and 78% (72–84) specificity, 83% (79–88) positive predictive value, and 54% (48–60) negative predictive value.21 In addition, 33% of the patients had an LVO, and the median DWI lesion volume was 2 mL in the alteplase group. Therefore, the results are limited to patients with small infarct volumes. The THAWS trial (Thrombolysis for Acute Wake-Up and Unclear Onset Strokes With Alteplase at 0.6 mg/kg) was terminated early after the publication of the WAKE-UP trial with 131 wake-up patients enrolled using the same DWI-FLAIR mismatch criteria as the WAKE-UP trial. The lower dose of alteplase failed to demonstrate benefit as compared to control for functional outcomes.22
With CT perfusion imaging selection, the EXTEND trial (Extending the time for Thrombolysis in Emergency Neurological Deficits) was performed in 225 patients (alteplase group 113 and placebo group 112). It determined that the alteplase group had a 44% greater likelihood to have minimal or no disability at 3 months when compared with the placebo group (adjusted risk ratio, 1.44 [95% CI, 1.01–2.06]) in patients with salvageable ischemic tissue within 9 hours of symptom onset. The criteria used to identify salvageable tissue on CT perfusion imaging was a mismatch ratio >1.2, absolute mismatch volume >10 mL, and ischemic core volume <70 mL. The sICH rate was higher in the alteplase group than the placebo group (6.2% versus 0.9%, P=0.05).23 A majority of the randomized patients were wake-up strokes (65%) and 6 to 9 hours from last known well before randomization (25%). Seventy percent of the patients had an LVO. The trial excluded lacunar infarct patients, and the premature termination of recruitment at 73% of the planned sample size affected the power of the study. Another prematurely terminated trial, the ECASS-4 (European Cooperative Acute Stroke Study 4) failed to show benefit of alteplase over placebo within 9 hours of symptom onset in 119 patients using MRI for patient selection with a perfusion-weighted imaging volume to ischemic core on DWI ratio of at least 1.2 and a perfusion lesion minimum volume of 20 mL. The early stopping of the trial with only 44% of sample size randomized due to slow recruitment significantly reduced the statistical power.24
The meta-analysis of EXTEND, ECASS-4, The EPITHET (Echoplanar Imaging Thrombolytic Evaluation Trial) demonstrated a clear benefit across all mRS outcome assessments (ordinal, mRS score of 0–1, mRS score of 0–2, particularly when automated perfusion mismatch criteria were met.25 In addition, a recent patient-level meta-analysis including 843 patients’ from the WAKE-UP,20 THAWS,22 EXTEND,23 and the ECASS-424 trials demonstrated that in patients who have had a stroke with an unknown time of onset with a DWI-FLAIR or CT perfusion mismatch, alteplase resulted in a better functional outcome at 90 days than placebo or standard care (adjusted OR, 1.49 [95% CI, 1.10–2.03]), despite an increased risk of sICH (3% versus <1%, P=0.024).26 Considering that most of these trials were terminated prematurely, more thrombolytic trials with a larger sample size and LVO as well as more distal occlusions are warranted in the late time window. Using the same imaging mismatch criteria as the EXTEND trial, an ongoing Chinese trial, Treatment With Intravenous Alteplase in Ischemic Stroke Patients With Onset Time Between 4.5 and 24 Hours is exploring the safety and efficacy of alteplase plus standard medical treatment versus standard medical treatment in the late window up to 24 hours after symptom onset (https://www.clinicaltrials.gov; Unique identifier: NCT04879615).

New Thrombolytic Drugs

TNK (tenecteplase) is generating a lot of interest in the stroke thrombolysis area and studies are striving to test whether it can be an alternative for or even can replace alteplase (Table 1 and Figure 3). TNK is a genetically engineered mutant tissue plasminogen activator with a longer half-life, greater fibrin specificity, and tolerance to plasminogen activator inhibitor-1 than alteplase, which allows for a bolus injection and does not require a 1-hour infusion.27
Table 1. Summary of the TNK Trials Since 2015
Study acronym; author; year publishedAim of study; study type; study size (N)Patient populationStudy intervention (no. of patients) /study comparator (no. of patients)Primary end point results (absolute event rates, P values; OR or RR; and 95% CI)
TEMPO-1; Coutts et al28; 2015Study aim: To assess the safety and feasibility of the use of 2 doses of TNK-tPA for the treatment of minor stroke with intracranial occlusion Study type: multicenter, prospective, uncontrolled, 2-cohort, TNK-tPA dose-escalation, safety, and feasibility trial Study size: 50Inclusion criteria: Acute ischemic stroke in an adult patient (aged, ≥18 y); onset (last-seen-well) time to treatment time <12 h; minor stroke defined as a baseline NIHSS <6 at the time of randomization; any acute intracranial occlusion (MCA, ACA, PCA, and vertebral arteries and basilar arteries) defined by CTA; prestroke independent functional status in activities of daily living with prestroke estimated modified Barthel Index of ≥90 and premorbid mRS score of 0–1; informed consent; patients can be treated within 90 min of the CT/CTA being completed.Intervention: Intravenous tenecteplase at 0.25 mg/kg (n=25) Comparator: Intravenous tenecteplase at 0.1 mg/kg (n=25)Primary end point: There were no drug-related serious adverse events in tier 1. In tier 2, there was 1 symptomatic intracranial hemorrhage (4%; 95% CI, 0.01–20.0).
ATTEST; Huang et al29; 2015Study aim: To assess the efficacy and safety of tenecteplase vs alteplase within 4·5 h of stroke onset in a population not selected on the basis of advanced neuroimaging, and to use imaging biomarkers to inform the design of a definitive phase 3 clinical trial Study type: Single-center, phase 2, prospective, randomized, open-label, blinded end point study. Study size: 104Inclusion criteria: 18 y or older; diagnosed supratentorial acute ischemic stroke with measurable deficit on NIHSS; within 4·5 h of symptom onset; living independently prestroke; considered eligible for intravenous thrombolysis according to clinical guidelinesIntervention: Tenecteplase (0.25 mg per kg, to a maximum 25 mg as a single bolus; n=52) Comparator: Alteplase (0·9 mg per kg to a maximum 90 mg, with 10% of dose as initial bolus, followed by 90% in a 1 h infusion; n=52)Primary end point: Of 71 patients (35 assigned tenecteplase and 36 assigned alteplase) contributing to the primary end point, no significant differences were noted for percentage of penumbral salvaged (68% [SD 28] for the tenecteplase group vs 68% [SD 23] for the alteplase group; mean difference 1·3% [95% CI, –9·6 to 12·1]; P=0.81). Safety end point: Neither incidence of symptomatic intracerebral hemorrhage (by SITS-MOST definition, 1/52 [2%] tenecteplase vs 2/51 [4%] alteplase, P=0.55; by ECASS II definition, 3/52 [6%] vs 4/51 [8%]; P=0.59) nor total intracerebral hemorrhage events (8/52 [15%] vs 14/51 [29%]; P=0.091) differed significantly between groups.
NOR-TEST; Logallo et al30; 2017Study aim: To investigate the safety and efficacy of tenecteplase vs alteplase in patients with acute stroke who were eligible for intravenous thrombolysis Study type: Phase 3, multicenter, prospective, randomized, open-label, blinded end point, superiority trial Study size: 1100Inclusion criteria: 18 y or older; clinically suspected acute ischemic stroke with measurable deficits on NIHSS; admitted within 4·5 h of symptom onset or within 4·5 h of awakening with symptoms; living independently prestroke; eligible for intravenous thrombolysis according to Norwegian clinical guidelines (off-label if mismatch between DW-MRI and FLAIR-MRI was detected)Intervention: Tenecteplase (0·4 mg/kg to a maximum of 40 mg as a single bolus intravenously; n=549) Comparator: Alteplase (0·9 mg/kg to a maximum of 90 mg, with 10% of the dose as initial bolus, followed by 90% in a 1 h intravenous infusion; n=551)Primary end point: Excellent functional outcome defined as mRS score 0–1 at 3 mo was achieved by 354 (64%) patients in the tenecteplase group and 345 (63%) patients in the alteplase group (OR 1·08 [95% CI, 0·84–1·38]; P=0.52). Safety end point: By 3 mo, there were no differences for any intracranial hemorrhage at 24–48 h, symptomatic intracranial hemorrhage at 24–48 h, or death between the two treatment groups in the per-protocol analysis.
EXTEND-IA TNK; Campbell et al36; 2018Study aim: To compare tenecteplase with alteplase in establishing reperfusion in patients before endovascular thrombectomy when it was administered within 4.5 h after the onset of symptoms Study type: Investigator-initiated, noninferiority followed by superiority, multicenter, prospective, randomized, open-label, blinded outcome trial Study size: 202Inclusion criteria: Could undergo intravenous thrombolysis within 4.5 h after the onset of ischemic stroke; had cerebral vascular occlusion on CT angiography of the internal carotid artery, the first segment of the middle cerebral artery, the second segment of the middle cerebral artery, or the basilar artery and if treatment to retrieve the intra-arterial clot could commence (arterial puncture) within 6 h after stroke onset (the criteria of CT perfusion mismatch were removed on October 12, 2016)Intervention: Tenecteplase (at a dose of 0.25 mg per kilogram of body weight; maximum dose, 25 mg; n=101) Comparator: Alteplase (at a dose of 0.9 mg per kilogram; maximum dose, 90 mg; n=101)Primary end point: Reperfusion of greater than 50% of the involved ischemic territory or an absence of retrievable thrombus at the time of the initial angiographic assessment occurred in 22% of the patients treated with tenecteplase vs 10% of those treated with alteplase (incidence difference, 12 percentage points; [95% CI, 2–21]; incidence ratio, 2.2; [95% CI, 1.1–4.4]; P=0.002 for noninferiority; P=0.03 for superiority). Safety end point: Symptomatic intracerebral hemorrhage occurred in 1% of the patients in each group.
EXTEND-IA TNK Part 2; Campbell et al31; 2020Study aim: To determine whether 0.40 mg/kg of tenecteplase safely improves reperfusion before endovascular thrombectomy vs 0.25 mg/kg of tenecteplase in patients with LVO ischemic stroke Study type: Investigator-initiated, multicenter, randomized, open-label, blinded end point trial Study size: 300Inclusion criteria: Adults; could receive intravenous thrombolysis within 4.5 h of ischemic stroke onset; had cerebral vascular occlusion on CTP of the intracranial internal carotid artery, middle cerebral artery first or second segments, or basilar artery and if endovascular thrombectomy was intended to be performedIntervention: Tenecteplase at 0.40 mg/kg (maximum, 40 mg) given as a bolus before endovascular thrombectomy (n=150) Comparator: Tenecteplase at 0.25 mg/kg (maximum, 25 mg) given as a bolus before endovascular thrombectomy (n=150)Primary end point: The number of participants with greater than 50% reperfusion of the previously occluded vascular territory was 29 of 150 (19.3%) in the 0.40 mg/kg group vs 29 of 150 (19.3%) in the 0.25 mg/kg group (unadjusted risk difference, 0.0% [95% CI, −8.9% to −8.9%]; adjusted risk ratio, 1.03 [95% CI, 0.66–1.61]; P=0.89). Safety end point: There were no significant differences between the 0.40 mg/kg and 0.25 mg/kg groups in all-cause deaths (26 [17%] vs 22 [15%]; unadjusted risk difference, 2.7% [95% CI, −5.6% to 11.0%]) or symptomatic intracranial hemorrhage (7 [4.7%] vs 2 [1.3%]; unadjusted risk difference, 3.3% [95% CI, −0.5% to 7.2%]).
ACA indicates anterior cerebral artery; ATTEST, Alteplase Versus Tenecteplase for Thrombolysis After Ischemic Stroke; CT, computed tomography; CTA, CT angiography; CTP‚ CT perfusion; DW, diffusion-weighted; ECASS, European Cooperative Acute Stroke Study; EXTEND-IA TNK part 2, Effect of Intravenous Tenecteplase Dose on Cerebral Reperfusion Before Thrombectomy in Patients With Large Vessel Occlusion Ischemic Stroke: The EXTEND-IA TNK Part 2 Randomized Clinical Trial; EXTEND-IA TNK, Tenecteplase Versus Alteplase Before Thrombectomy for Ischemic Stroke; FLAIR, fluid-attenuated inversion recovery; LVO, large vessel occlusion; MCA, middle cerebral artery; MRI, magnetic resonance imaging; NIHSS, National Institutes of Health Stroke Scale; NOR-TEST, Tenecteplase Versus Alteplase for Management of Acute Ischaemic Stroke; PCA, posterior cerebral artery; TEMPO-1, Tenecteplase-Tissue-Type Plasminogen Activator Evaluation for Minor Ischemic Stroke With Proven Occlusion; TNK, tenecteplase; and tPA, tissue-type plasminogen activator.
Figure 3. History of tenecteplase trials in acute ischemic stroke from 2015. AcT indicates Alteplase Compared to Tenecteplase in Patients With Acute Ischemic Stroke; ATTEST, Alteplase Versus Tenecteplase for Thrombolysis After Ischemic Stroke; BRETIS-TNK, Boosting recanalization of thrombectomy for ischemic stroke by intra-arterial TNK; CHABLIS-T, Chinese Acute Tissue-Based Imaging Selection for Lysis In Stroke-Tenecteplase; ETERNAL-LVO, Extending the Time Window for Tenecteplase by Effective Reperfusion in Patients With LVO; EXTEND-IA TNK part 2, Effect of Intravenous Tenecteplase Dose on Cerebral Reperfusion Before Thrombectomy in Patients With Large Vessel Occlusion Ischemic Stroke: The EXTEND-IA TNK Part 2 Randomized Clinical Trial; EXTEND-IA TNK, Tenecteplase Versus Alteplase Before Thrombectomy for Ischemic Stroke; LVO, large vessel occlusion; INSIST-TNK, Improving neuroprotective strategy for ischemic stroke with poor recanalization after thrombectomy by intra-arterial TNK; NOR-TEST, Tenecteplase Versus Alteplase for Management of Acute Ischaemic Stroke; ROSE-TNK, MRI-Guided Thrombolysis for Stroke Beyond Time Window by TNK; TASTE, Tenecteplase versus Alteplase for Stroke Thrombolysis Evaluation; Australian New Zealand; TEMPO-1, Tenecteplase-Tissue-Type Plasminogen Activator Evaluation for Minor Ischemic Stroke With Proven Occlusion; TIMELESS, Tenecteplase in Stroke Patients Between 4.5 and 24 Hours; and TWIST, Tenecteplase in Wake-Up Ischemic Stroke Trial.
In 2005, a pilot dose-escalation safety study found that TNK doses of 0.1 to 0.4 mg/kg were safe in ischemic stroke.32 Then the Australian TNK (low-dose tenecteplase versus standard-dose alteplase for acute ischemic stroke) trial in 201233 demonstrated that TNK 0.25 mg/kg was superior to TNK 0.1 mg/kg and alteplase 0.9 mg/kg for recanalization and 24-hour clinical improvement in 75 total patients with a perfusion imaging mismatch and a proximal vessel occlusion. Although the sample size was very small, it was the first trial to test the optimal dose for TNK. The ATTEST phase 2 trial (Alteplase-Tenecteplase Trial Evaluation for Stroke Thrombolysis),29 which enrolled 104 patients, found a similar percentage of penumbral salvage on CT perfusion when comparing TNK and alteplase (68% versus 68%) when administered within 4.5 hours of stroke onset.
A pooled analysis of these 2 trials showed that TNK with LVO had a significantly better rate of complete vessel recanalization at 24 hours (71% versus 43%, P<0.001), early clinical improvement, and higher rates of mRS score of 0 to 1 at 90 days (OR, 4.82 [95% CI, 1.02–7.84]; P=0.05).34 The benefits of TNK were possibly more prominent in patients with a defined target mismatch (absolute mismatch volume >15 mL, mismatch ratio >1.8, baseline ischemic core <70 mL, and volume of severely hypoperfused tissue <100 mL).35
The contemporary TEMPO-1 study (TNK-tPA Evaluation for Minor Ischemic Stroke With Proven Occlusion)28 also confirmed that the complete recanalization rate was higher with the dose of 0.25 mg/kg than that observed with 0.1 mg/kg (52% versus 39%) in 50 patients with minor stroke (National Institutes of Health Stroke Scale [NIHSS] score of ≤5) and intracranial occlusion within 12 hours of symptom onset. However, the study was a proof-of-concept safety study in a small sample that did not compare TNK to a matched control group of patients who used standard antiplatelet treatment or alteplase. The ongoing TEMPO-2 (A Randomized Controlled Trial of TNK-tPA Versus Standard of Care for Minor Ischemic Stroke With Proven Occlusion) (https://www.clinicaltrials.gov; Unique identifier: NCT02398656) TNK-tPA is comparing TNK 0.25 mg/kg with antiplatelet agent(s) in minor stroke patients with LVO within 12 hours of symptom onset, targeting a sample size of 1274.
The NOR-TEST (Norwegian Tenecteplase Stroke Trial),30 the first phase 3 clinical trial of TNK at a high dose (0.4 mg/kg) versus alteplase in 1100 patients who fulfilled standard thrombolysis eligibility criteria failed to show superiority of TNK over alteplase (64% versus 63%, respectively) rates of excellent outcome (mRS score of 0–1) at 3 months (OR, 1.08 [95% CI, 0.84–1.38]). The frequency of sICH (2%–3%) was similar between groups. The trial included a majority of mild stroke patients and 17% had stroke mimics, which diluted the efficacy of TNK over alteplase. The ongoing NOR-TEST 2 (https://www.clinicaltrials.gov; Unique identifier: NCT03854500) is enrolling patients with NIHSS >5 to investigate the efficacy and safety of TNK 0.4 mg/kg versus alteplase within 4.5 hours after symptom onset or wake-up stroke patients with a DWI-FLAIR mismatch.
It is uncertain if 0.25 mg/kg is the optimal dose for TNK for bridging therapy. The EXTEND-IA TNK study (Tenecteplase Versus Alteplase Before Thrombectomy for Ischemic Stroke)36 confirmed that TNK 0.25 mg/kg significantly increased by an absolute 12% rate successful recanalization before thrombectomy as compared to alteplase in patients with LVO who were thrombectomy candidates within 4.5 hours of stroke onset. The subsequent EXTEND-IA TNK part 2 (Effect of Intravenous Tenecteplase Dose on Cerebral Reperfusion Before Thrombectomy in Patients With Large Vessel Occlusion Ischemic Stroke: The EXTEND-IA TNK Part 2 Randomized Clinical Trial)31 showed a similar percentage (19.3%) in >50% reperfusion of the previously occluded vascular territory comparing TNK doses of 0.4 mg/kg and 0.25 mg/kg. There were no significant differences in all-cause deaths and sICH between the two groups. These trials pave the way for phase 3 trials to determine whether TNK 0.25 mg/kg is superior to alteplase regarding functional outcomes. A recent meta-analysis of TNK in patients with LVO showed that TNK demonstrated successful recanalization (OR, 3.05 [95% CI, 1.73–5.40]), higher rates of mRS 0 to 2 (OR, 2.06 [95% CI, 1.15–3.69]), and functional improvement (OR, 1.84 [95% CI, 1.18–2.87]) at 3 months compared to alteplase.37 Therefore, TNK may serve as an alternative for alteplase in acute ischemic stroke, and further active comparator trials are warranted to test its efficacy and safety for functional outcomes. Ongoing phase 3 trials, including TASTE (Tenecteplase Versus Alteplase for Stroke Thrombolysis Evaluation; Australian New Zealand; https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=363714&isReview=true; ACTRN12613000243718), ATTEST2 (https://www.clinicaltrials.gov; Unique identifier: NCT02814409), and AcT (Alteplase Compared to Tenecteplase in Patients With Acute Ischemic Stroke; https://www.clinicaltrials.gov; Unique identifier: NCT03889249) may provide more evidence for the efficacy of TNK 0.25 mg/kg in acute ischemic stroke within 4.5 hours of symptom onset.
With imaging selection, several trials are exploring the efficacy and safety of TNK in the extended time window beyond 4.5 hours after symptom onset. The ongoing TWIST (Tenecteplase in Wake-Up Ischemic Stroke Trial, https://www.clinicaltrials.gov; Unique identifier: NCT03181360), CHABLIS-T (Chinese Acute Tissue-Based Imaging Selection for Lysis In Stroke-Tenecteplase, https://www.clinicaltrials.gov; Unique identifier: NCT04086147), TIMELESS (Tenecteplase in Stroke Patients Between 4.5 and 24 Hours, https://www.clinicaltrials.gov; Unique identifier: NCT03785678), ETERNAL-LVO (Extending the Time Window for Tenecteplase by Effective Reperfusion in Patients With LVO, https://www.clinicaltrials.gov; Unique identifier: NCT04454788), ROSE-TNK (MRI-Guided Thrombolysis for Stroke Beyond Time Window by TNK, https://www.clinicaltrials.gov; Unique identifier: NCT04752631) are exploring the efficacy of tenecteplase in the late time window (4.5–24 hours, wake-up stroke or unknown onset time).
Based on the current limited evidence, the 2019 American Heart Association/American Stroke Association acute stroke guidelines recommended that it may be reasonable to choose TNK (single IV bolus of 0.25 mg/kg, maximum dose 25 mg) over alteplase in patients without contraindications for thrombolysis who are also eligible to undergo mechanical thrombectomy. TNK 0.4 mg/kg might be considered as an alternative to alteplase in patients with minor neurological impairment and no major intracranial occlusion (IIb class of recommendation, Level B (moderate-quality) of evidence from randomized clinical trials [B-R] level of evidence).38 Currently, TNK is Food and Drug Administration approved for the treatment of myocardial ischemia and is under further investigation for the treatment of acute ischemic stroke.

Other Thrombolytic Drugs

Staphylokinase was first isolated and purified in 1948. It has high fibrin-selectivity, but early experience in patients with acute myocardial infarction showed that many patients developed neutralizing antibodies, preventing further research on this agent.39 A new thrombolytic drug, nonimmunogenic staphylokinase is a modified recombinant staphylokinase with low immunogenicity, high thrombolytic activity, and selectivity to fibrin. Recently, the FRIDA trial (Nonimmunogenic Recombinant Staphylokinase Versus Alteplase for Patients With Acute Ischemic Stroke 4.5 Hours After Symptom Onset in Russia) in 385 patients revealed that nonimmunogenic staphylokinase was noninferior to alteplase on favorable functional outcome (OR, 1.47 [95% CI, 0.93–2.32) with comparable sICH rates.40 This phase 3, randomized, multicenter, parallel-group, open-label, noninferiority trial was challenged because of its relatively wide noninferiority margin (16%), small sample size, and lack of phase 2 trial data in acute ischemic stroke.41 Further trials are needed to investigate the superiority of nonimmunogenic staphylokinase compared with alteplase.

Alteplase in Minor Stroke

The European Stroke Organization guidelines on intravenous thrombolysis for acute ischemic stroke recommended not giving intravenous thrombolysis for patients with acute minor nondisabling ischemic stroke of <4.5-hour duration based on the PRISMS trial (The Potential of rtPA for Ischemic Strokes With Mild Symptoms)42,43 The PRISMS trial enrolled 313 patients with minor neurological deficits (NIHSS score of 0–5) judged to not be clearly disabling within 3 hours of stroke onset, randomized to intravenous alteplase or aspirin arms and failed to show superiority of alteplase over aspirin.42 However, the PRISMS trials were prematurely terminated and no robust conclusions can be drawn from it. In addition, dual antiplatelet therapy was already recommended for minor stroke patients44; therefore, further trials are needed to compare alteplase with dual antiplatelets for minor stroke patients. Currently, evidence is lacking about alteplase use for patients with acute minor nondisabling ischemic stroke of <4.5-hour duration, and with proven LVO. The TEMPO-2 trial as mentioned previously is testing the efficacy and safety of tenecteplase versus antiplatelets in patients with minor stroke and LVO (https://www.clinicaltrials.gov; Unique identifier: NCT02398656).
Recommendations for intravenous thrombolysis
tPA is the treatment of choice in 4.5 hour time window unless there are contraindications to its use.
Intravenous tPA should be considered in carefully selected patients using advanced imaging beyond 4.5 hours.
TNK can be considered as alternative treatment but is currently not Food and Drug Administration approved for ischemic stroke.
Treatment should be initiated as quickly as possible, potentially with the use of MSUs when available.
Intravenous tPA is of uncertain value in patients with minor stroke and additional clinical trials are needed.

Endovascular Revascularization (Thrombectomy/Thrombaspiration)

A meta-analysis of 5 major RCTs (MR CLEAN [Multicenter Randomized Clinical Trial of Endovascular Treatment for Acute Ischemic Stroke in the Netherlands],45 ESCAPE [Endovascular Treatment for Small Core and Anterior Circulation Proximal Occlusion With Emphasis on Minimizing CT to Recanalization Times],46 REVASCAT [Randomized Trial of Revascularization With Solitaire FR Device Versus Best Medical Therapy in the Treatment of Acute Stroke Due to Anterior Circulation Large Vessel Occlusion Presenting Within Eight Hours of Symptom Onset],47 SWIFT PRIME [Solitaire With the Intention for Thrombectomy as Primary Endovascular Treatment],48 and EXTEND-IA [Extending the Time for Thrombolysis in Emergency Neurological Deficits—Intra-Arterial]49) showed that EVT was superior to medical management of acute ischemic stroke associated with LVO of the middle cerebral artery (MCA) and internal carotid artery (ICA) primarily within 6 hours of symptom onset as seen in Figure 4.9 EVT led to significantly reduced disability at 90 days compared with control (adjusted common OR, 2·49 [95% CI, 1·76–3·53]; P<0.0001). The number of patients needed to treat to reduce disability by at least one level on the mRS for one patient was 2.6. EVT was beneficial over medical management in several prespecified subgroups, including in subjects ≥80 years (common OR, 3.68 [95% CI, 1.95–6.92]), patients randomized >5 hours after symptom onset (1·76, 1·05–2·97), and those not qualifying for intravenous tPA (2·43, 1·30–4·55; mixed-effects ordinal logistic regression models). Mortality at 90 days and the risk of sICH did not differ between studied populations.
Figure 4. Endovascular treatment for acute ischemic stroke associated with large vessel occlusion. A, Computed tomography (CT) angiographic study shows a partial occlusion of left M1/middle cerebral artery (MCA) segment and total occlusion of superior M2 division (arrows). B and C, CT shows hyperdensity of lentiform nucleus (arrow) corresponding to infarcted brain tissue, Alberta Stroke Program Early CT Score 9. D, CT perfusion scan shows no core infarct (CBF <30%) and a large penumbra (green area) with a mismatch volume of 85 mL. E–G, Left common carotid artery angiogram shows a dissection and subsequent occlusion of left internal carotid artery (ICA, arrow) and partial thromboembolic occlusion of left terminal ICA and clot extension into M1/MCA (arrow), inferior and superior M2 divisions as well as anterior cerebral artery with a Treatment in Cerebral Ischemia Score (TICI)=1 flow. Thrombaspiration catheter in place (arrow). H and I, Acute carotid artery stenting and contact thrombaspiration resulting in a complete revascularization and a TICI=3 flow. H, Follow-up fluid-attenuated inversion recovery–magnetic resonance imaging shows only a small ischemic area in left lentiform nucleus and insula (arrows).
Subsequent EVT studies, the DAWN (Diffusion-Weighted Imaging or Computerized Tomography Perfusion Assessment with Clinical Mismatch in the Triage of Wake-Up and Late Presenting Strokes Undergoing Neurointervention With Trevo)50 and DEFUSE 3 trials (Endovascular Therapy Following Imaging Evaluation for Ischemic Stroke)51 extended the time window for revascularization up to 24 hours after symptom onset. DAWN compared the use of a specific stent-retriever (Trevo, Stryker NV, Fremont, CA) for a 6- to 24-hour time window.50 Patients had to have a mismatch between the severity of the clinical deficit and the infarct volume associated with an ICA or M1/MCA occlusion. Infarct volume was determined with diffusion-weighted MRI or perfusion CT (automated software RAPID, iSchemaView). A total of 107 subjects were randomized to EVT and 99 to the control group. Median infarct volume was small in both subgroups (7.6 mL for EVT and 8.9 mL for control group; interquartile range, 2.0–18.0 and 3.0–18.1). The rate of functional independence at 90 days was 49% in the thrombectomy group as compared to 13% in the control group (adjusted difference, 33% [95% CI, 21–44]; posterior probability of superiority, >0.999). The rate of neurological deterioration was lower in the EVT group than in the control group (14% versus 26%; absolute difference, −12 percentage points; 95% CI, −23 to −1; P=0.04). Serious adverse event rates including stroke-related death at 90 days and sICH did not differ between the two subgroups. The DEFUSE 3 trial enrolled patients within the 6- to 16-hour time window and an infarct size <70 mL who had a ratio of volume of ischemic tissue to initial infarct volume of 1.8 or more on CT perfusion or MR diffusion and perfusion imaging (RAPID software, iSchemaView).51 If needed, carotid angioplasty with or without stenting was permitted as part of the intervention. Based on an interim analysis, the trial was halted prematurely because the prespecified efficacy boundaries were reached. The rate of complete recanalization at 24 hours in the EVT group was significantly higher (78% versus 18% for medical therapy). A higher percentage of patients were functionally independent (0–2 mRS) following EVT as compared to the medical arm (45% versus 17%, risk ratio, 2.67 [95% CI, 1.60–4.48]; P<0.001). The 90-day mortality rate was 14% in the EVT group versus 26% in the control arm (P=0.05), the frequency of sICH (7% and 4%, P=0.75), and serious adverse events (43% and 53%, P=0.18). Interestingly, despite the clinical benefit of EVT, there was no difference in median infarct growth and volume at 24-hour as compared to medical therapy. The investigators suggest that infarct growth may develop over several days in patients who do not experience reperfusion. Both trials expanded the time window for EVT up to 24 h based on CT/MR-perfusion imaging and a mismatch in a selected patient population. In 2019, the American Heart Association/American Stroke Association acute stroke guidelines expanded the class I level A recommendation for LVO of MCA and ICA up to 16 hours and class IIA level B-R recommendation for patients presenting between 16- and 24-hour time window and a NIHSS score of ≥6.38 However, the recently published AURORA (Analysis of pooled data from randomized studies of thrombectomy more than 6 hours after last known well) meta-analysis of randomized controlled EVT trials >6 hours after last-well-known, suggests that the American Heart Association guideline distinction between DEFUSE 3 (6–16 hours) and DAWN (6–24 hours) selection criteria is unnecessary.52 Thrombectomy was beneficial over best medical management with an unadjusted common OR of 2.42 (95% CI, 1·76–3.33]; P<0·0001) and higher rates of independent living (mRS score ≤2; 122 [45.9%] of 266 versus 46 [19.3%] of 238; P<0.0001). No heterogeneity of treatment effect was found across subgroups characterized by age, gender, baseline stroke severity, arterial occlusion site, baseline Alberta Stroke Program Early CT Score, and mode of presentation. The EVT treatment effect was stronger in patients randomly assigned within 12 to 24 hours (common OR, 5.86 [95% CI, 3.14–10.94]) than those randomized between 6 and 12 hours (OR, 1.76 [95% CI, 1.18–2.62]; P for interaction=0.0087). At 90-day no significant difference in mortality or symptomatic ICH was found between the intervention and control groups (mortality 44 [16·5%] of 266 versus 46 [19·3%] of 238; sICH 14 [5·3%] of 266 versus 8 [3·3%] of 239). Pooled mixed-effects modeling with fixed effects for parameters of interest showed that EVT was associated with a reduction in disability at 90 days in the clinical mismatch (OR, 3.57 [95% CI, 2.29–5.57]; P<0.001) and target perfusion mismatch (OR, 3.13 [95% CI, 2.10–4.66]; P=0.001) subgroups as compared with standard medical management in patients presenting 6- to 24-hour last known well.53 The best OR calculated was for the subgroup between 12 and 24 hours after last known well (clinical mismatch subgroup, OR, 4.95 [95% CI, 2.20–11.16]; P<0.001; target perfusion mismatch subgroup, OR, 5.01 [95% CI, 2.37–10.60]; P<0.001).
A recently published multinational cohort study showed that in the extended time window (within 6 to 24 hours last known well) there were no significant differences in 90-day clinical outcomes (ordinal mRS shift) when patients for EVT were selected based on a noncontrast CT compared with those selected with CTP or MRI (CT versus CTP, adjusted OR, 0.95 [95% CI, 0.77–1.17]; P=0.64), CT versus MRI adjusted OR, 0.95 [95% CI, 0.8–1.13]; P=0.55) but lower in patients selected by MRI than CT (adjusted OR, 0.79 [95% CI, 0.64–0.98]; P=0.03). Successful reperfusion was more common in the CT and CTP groups compared with the MRI group (474 [88.9%] and 670 [89.5%] versus 250 [78.9%]; P<0.001). No significant differences in symptomatic ICH (CT, 42 [8.1%]; CTP, 43 [5.8%]; MRI, 15 [4.7%]; P=0.11) or 90-day mortality (CT, 125 [23.4%]; CTP, 159 [21.1%]; MRI, 62 [19.5%]; P=0.38) were found.54 Thus, in this patient population the use of noncontrast CT, readily available in most centers, may suffice to make the decision for an EVT treatment.
EVT trials (DIRECT ANGIO [Effect of Direct Transfer to Angiosuite on Functional Outcome in Patient With Severe Acute Stroke Treated With Thrombectomy, https://www.clinicaltrials.gov; Unique identifier: NCT03969511],55 WE-TRUST [Workflow Optimization to Reduce Time to Endovascular Reperfusion for Ultra-fast Stroke Treatment, https://www.clinicaltrials.gov; Unique identifier: NCT04701684]) are underway to further improve the workflow and reduce time to treatment by direct patient transfer to catheter labs, bypassing the Emergency Department (ED) and conventional CT scanners. Newer angiography systems used for EVT have either integrated CT scanning capabilities using flat-panel detectors (Cone-beam CT) or provide a hybrid system composed of a CT scanner and an angiography system. Early but growing experience in randomized clinical trials has shown a 50%-time reduction in symptom-to-recanalization with direct transfer to the angiography suite and significant improvement in clinical outcomes.56–58 Several comprehensive stroke centers have already adopted this workflow successfully.
Alternative thrombectomy strategies have been developed as the first pass technique with direct aspiration being one of the most promising. In the ASTER trial (Contact Aspiration versus Stent Retriever for Successful Revascularization), first-line thrombectomy with contact aspiration was compared with stent-retriever. Contact aspiration did not result in an increased successful revascularization rate.59 For the primary outcome, successful revascularization was achieved in 85.4% in the contact aspiration group versus 83.1% in the stent-retriever group (OR, 1.20 [95% CI, 0.68–2.10]; P=0.53; difference 2.4% [95% CI, −5.4% to 9.7%]). There were no differences in clinical efficacy and serious adverse events between both groups. Similarly in the COMPASS trial (Aspiration Thrombectomy Versus Stent Retriever Thrombectomy as First-Line Approach for Large Vessel Occlusion) trial, although cost-effective, direct aspiration as first-pass thrombectomy showed a noninferior functional outcome at 90 days compared with stent-retriever first-line thrombectomy.60 A modified Rankin score of 0–2 at 90 days was achieved in 52% (95% CI, 43.8–60.3) in the aspiration group and versus 50% (95% CI, 41.6–57.4]) in the stent-retriever group, showing that aspiration as first pass was noninferior to stent-retriever first-line (noninferiority P=0.0014). There were no differences in ICH or all-cause mortality at 3 months between both groups. Of note, in 21% of the aspiration first-pass thrombectomy group, a stent-retriever was used. Distal aspiration during stent-retriever thrombectomy was used in 85% of the first-line stent-retriever group.
Critical for a good functional outcome (mRS score of 0–1) as adjudicated at 90 days, remains an early and complete revascularization (grade 3 on the Treatment in Cerebral Ischemia Score 3) achieved with a single pass.61 Clot characteristics, that is, density, prone to fracture, amount of calcification, perviousness, length, location, and vessel wall interaction may all play a role in achieving a first pass effect and is being studied using CT imaging and various prediction algorithms based on imaging.62,63
Use of balloon guide catheters to induce flow arrest and reversal during EVT has proven to reduce the time to revascularization, increase the first pass effect during thrombectomy and improve clinical outcome and was described initially based on the NASA (North American Solitaire Acute Stroke) registry.64 The EVT–balloon guide catheter group had a higher rate of first-pass effect (48%) versus EVT with use of a conventional guide catheter (26%; P=0.001) or EVT with a distal access catheter (35%; P=0.002).65 The balloon guide catheter group achieved the highest rate of functional independence (61%) versus conventional guide catheter (42%; P=0.007) and distal access catheter (52%; P=0.027). Final revascularization and mortality rates did not differ across the groups.
To broaden the indication for EVT, numerous ongoing trials are investigating the value of thrombectomy alone or in combination with thrombolysis for patients presenting within 4.5-hour time window, with a large core volume infarct >70 mL, an Alberta Stroke Program Early CT Score <6, mild strokes NIHSS score <6 and subjects presenting with distal anterior (A1/A2) and middle cerebral artery (M2/M3) occlusion. Based on a pooled meta-analysis of several EVT studies, the 2019 American Heart Association/American Stroke Association guidelines have issued a class IIB recommendation for EVT in patients with a symptomatic occlusion of M2 or M3/MCA segments.38,66 Limitations of these studies include small sample sizes, selection bias, definition and size of treated M2 segments, lack of control groups, and various EVT modalities used. Nevertheless, the 2019 Society for Neuro-Interventional Surgery guidelines have issued a class I, level A recommendation for thrombectomy in occlusions of the ICA (including intracranial, cervical segments, or tandem occlusion), and M1/M2-MCA that replaces the 2015 Society for Neuro-Interventional Surgery guidelines in which thrombectomy recommendations were limited to ICA and MCA M1 LVO locations.67 Although single-center case series have demonstrated the safety and feasibility of EVT for patients with a low NIHSS <6, a recent multicenter study of 251 subjects with LVO did not show any significant difference between the EVT and the medical arm.68 Other future studies will have to clarify the role of EVT in the growing population of nonagenarians presenting with acute ischemic stroke, subjects <18 years of age, patients with a ≥3 mRS at baseline, subjects with a large core stroke volume with a low Alberta Stroke Program Early CT Score, and impact of associated co-morbidities, for example, cancer on functional outcome.
Use of conscious sedation versus general anesthesia (GA) for EVT has been debated over the past decade but has not shown significant difference in clinical outcomes between both groups.69 A total of 150 subjects were studied. The mean NIHSS score for the GA group was 16.8 at admission versus 13.6 after 24 hours; difference, −3.2 points (95% CI −5.6 to −0.8), and for the conscious sedation group, the mean NIHSS score was 17.2 at admission versus 13.6 after 24 hours; difference, −3.6 points (95% CI −5.5 to −1.7); mean difference between groups, −0.4 (95% CI −3.4 to 2.7; P=0.82). However, in the GA versus conscious sedation group, a higher rate of hypothermia (32.9% versus 9.1%; P<0.001) and pneumonia (13.7% versus 3.9%; P=0.03) were encountered. There were no differences in mortality at 3 months (24.7% in both groups). With the exception of a recently published meta-analysis including randomized data from 3 single-center trials,70 other randomized studies have reached similar conclusions that GA did not impact clinical outcome as compared to conscious sedation.71 This study showed an increased risk for hypotension during GA (decline in systolic blood pressure of >20% from baseline (80.8% versus 53.1% for the conscious sedation subgroup; OR, 4.26 [95% CI, 2.55–7.09]; P<0.001) and blood pressure variability (systolic blood pressure >180 mm Hg or <120 mm Hg; 79.7 versus 62.3%; OR, 2.42 [95% CI, 1.49–3.93]; P<0.001) impacting the mean 90-day mRS score (2.8 [95% CI, 2.5–3.1]) in the general anesthesia group versus (3.2 [95% CI, 3.0–3.5]) in the sedation group (difference, 0.43 [95% CI, 0.03–0.83]; common OR, 1.58 [95% CI, 1.09–2.29]; P=0.02).70
Although effective in the anterior circulation, the value of EVT in basilar artery occlusion has not been established in RCTs. Two recently published studies (BEST [Basilar Artery Occlusion Endovascular Intervention Versus Standard Medical Treatment]72 and BASICS [Basilar Artery International Cooperation Study]73 trials) showed no benefit of EVT for basilar artery occlusion. The BEST trial after enrolling 131 patients in China was prematurely terminated because of a high crossover rate and poor recruitment. In the small sample size studied 42% of patients in the EVT and 32% in the medical arm had an mRS of 0 to 3 at 90 days (statistically not significant). However, patients treated with EVT per-protocol had significantly better outcomes for all the primary and secondary functional end points on adjusted analysis compared with those of patients receiving standard medical care. In the BASICS trial, EVT was initiated at a median of 4.4 hours after stroke onset, and 154 subjects were enrolled in the EVT group and 146 in the medical arm. There was no difference in the use of intravenous thrombolysis in both groups (78.6% in EVT and 79.5% in the medical group). A favorable functional outcome occurred in 44.2% of the EVT population versus 37.7% in the medical arm (risk ratio, 1.18 [95% CI, 0.92–1.50]). sICH occurred in 4.5% of the patients after endovascular therapy and in 0.7% of those after medical therapy (risk ratio, 6.9 [95% CI, 0.9–53.0]); mortality at 90 days was 38.3% and 43.2%, respectively (risk ratio, 0.87 [95% CI, 0.68–1.12]). The study has several limitations including that a large percentage of patients received EVT outside the trial thus causing selection bias.74 A beneficial impact of EVT in these subjects was not reported. Patients would have likely benefited from EVT with appropriate use of imaging, such as assessment of the collateral circulation on CT angiography and size of core infarct on MRI diffusion.75 As proposed by the editorial, a sensitive outcome measures of functional disability in patients with strokes in the basilar artery territory should be developed.74

Skipping Intravenous Thrombolysis and Direct to Thrombectomy

Intravenous thrombolysis with alteplase is a widely used treatment that is easily performed in a variety of hospital settings. It can potentially facilitate mechanical thrombectomy as well as increase the possibility of reperfusion when it is difficult for thrombectomy to access the entire thrombus (eg, due to excessively tortuous vessels). However, the downsides of alteplase include the possibility of causing intracerebral hemorrhage, thrombus migration, delaying groin puncture, and increasing the expense of treatment. Moreover, the recanalization rate with intravenous thrombolysis varies from 10% to 30% based on the vessel occlusion site and is particularly low in patients with a large thrombus burden in the distal ICA.76 Therefore, whether proceeding directly to thrombectomy is comparable to bridging therapy with the combination of intravenous thrombolysis and thrombectomy is an important question.
Four randomized controlled trials have been published, three of the trials were performed in Asian patients, and other one trial was in whites (Table 2). In all the trials, the time windows for randomization were within 4.5 hours after symptom onset. The first head-to-head comparison trial of bridging therapy and direct to thrombectomy was the DIRECT-MT (Direct Intraarterial Thrombectomy to Revascularize Acute Ischemic Stroke Patients With LVO Efficiently in Chinese Tertiary Hospitals: a Multicenter Randomized Clinical Trial). It randomly assigned 656 acute ischemic stroke patients with anterior circulation LVO to bridging therapy or direct to thrombectomy. Based on a generous inferiority margin OR, 0.8, the direct to thrombectomy group was noninferior to bridging therapy on the 90-day functional outcome (adjusted common OR, 1.07 [95% CI, 0.81–1.40]). The sICH and death rate were not significantly different between the two arms.79 The trial was challenged because of its generous noninferiority margin, complicated prehospital triage system, and longer workflow times. Consistent with the DIRECT-MT, the DEVT trial (Direct Endovascular Thrombectomy Versus Combined IVT and Endovascular Thrombectomy for Patients With Acute LVO in the Anterior Circulation) involving 234 Chinese anterior circulation LVO patients revealed that the direct to thrombectomy group achieved a similar rate of functional independence compared with the bridging therapy group at the 90-day follow-up (difference, 7.7% [1-sided 97.5% CI, −5.1% to ∞]; P for noninferiority=0.003).77 The study used 10% for the favorable outcome proportion difference as the noninferiority margin, which was much wider than the minimal clinically important differences 3.5% to 4.4% of the functional independence proportion that was recommended by stroke expert survey studies for acute ischemic stroke.81 In Japan, the SKIP (Direct Mechanical Thrombectomy in Acute LVO Stroke) trial in 204 acute ischemic stroke patients due to anterior circulation LVO failed to show noninferiority of direct to thrombectomy as compared to bridging therapy using a reduced dose of alteplase (0.6 mg/kg). In addition, the noninferiority margin of 0.74 in this study significantly underestimated the sample size.78 A meta-analysis of these 3 trials comprising 1,092 Asian patients found no difference between the direct to thrombectomy and bridging therapy groups on the 90-day mRS score of 0 to 2 (adjusted OR 1.11, 95% CI 0.76 to1.63), mRS score of 0 to 1 (adjusted OR, 1.16 [95% CI, 0.84–1.61]), and functional improvement at 3 months (adjusted common OR, 1.09 [95% CI, 0.86–1.37]).82
Table 2. RCTs Comparing the Effect of EVT Alone and Intravenous Thrombolysis Followed by EVT
Study acronym; author; year publishedAim of study; study type; study size (N)Patient PopulationStudy Intervention (no. of patients)/study comparator (no. of patients)Primary end point results (absolute event rates, P values; OR or RR; and 95% CI)
DEVT; Zi et al77; 2021Study aim: To investigate whether endovascular thrombectomy alone is noninferior to intravenous alteplase followed by endovascular thrombectomy Study type: Multicenter, randomized, noninferiority trial Study size: 234Inclusion criteria: 18 y or older with acute ischemic stroke, eligible for IV alteplase treatment within 4.5 h of onset, and had occlusion of the intracranial internal carotid artery or the first segment of the middle cerebral artery confirmed by CTA or MRAIntervention: Endovascular thrombectomy alone group(n=116) Comparator: Combined intravenous thrombolysis and endovascular thrombectomy group (n=118)Primary end point: Stopped early because of noninferiority. At the 90-day follow-up, 63 patients (54.3%) in the endovascular thrombectomy alone group vs 55 (46.6%) in the combined treatment group achieved functional independence at the 90-day follow-up (difference 7.7%, 1-sided 97.5% CI −5.1% to ∞), P for noninferiority=0.003). Safety end point: No significant between-group differences were detected in symptomatic intracerebral hemorrhage and 90-day mortality.
SKIP; Suzuki et al78; 2021Study aim: To examine whether mechanical thrombectomy alone is noninferior to combined intravenous thrombolysis plus mechanical thrombectomy for favorable poststroke outcome. Study type: Investigator-initiated, multicenter, randomized, open-label, noninferiority clinical trial Study size: 204Inclusion criteria: 18 to 85 y old; had acute stroke with internal carotid artery (ICA) or M1 occlusion evaluated by MRA or CTA; had a baseline ASPECTS of 6 to 10 or DWI-ASPECTS of 5 to 10; initial NIHSS score equal to 6 or greater; were functionally independent before stroke, with mRS score of 0 to 2; met the criteria of the Japanese guidelines for treatment with the lower dose of 0.6 mg/kg of alteplase as intravenous thrombolysis within 4.5 h from onset.Intervention: Mechanical thrombectomy alone (n=101) Comparator: Combined intravenous thrombolysis (alteplase at a 0.6 mg/kg dose) plus mechanical thrombectomy (n=103).Primary end point: Favorable outcome occurred in 60 patients (59.4%) in the mechanical thrombectomy alone group and 59 patients (57.3%) in the combined intravenous thrombolysis plus mechanical thrombectomy group, with no significant between-group difference (difference 2.1% [1-sided 97.5% CI, −11.4% to∞]; OR 1.09 [1-sided 97.5% CI, 0.63 to ∞]; P=0.18 for noninferiority). Safety end point: Any intracerebral hemorrhage was observed less frequently in the mechanical thrombectomy alone group than in the combined group (34 [33.7%] vs 52 [50.5%]; difference –16.8% [95% CI, –32.1% to –1.6%]; OR 0.50 [95% CI, 0.28–0.88]; P=0.02). Symptomatic intracerebral hemorrhage was not significantly different between groups (6 [5.9%] vs 8 [7.7%]; difference –1.8% [95% CI, –9.7% to 6.1%]; OR 0.75 [95% CI, 0.25–2.24]; P=0.78).
DIRECT-MT; Yang et al79; 2020Study Aim: To investigate the benefit and risk of administering intravenous alteplase before endovascular thrombectomy. Study type: Investigator-initiated, multicenter, prospective, open-label RCT with blinded outcome assessment Study size: 656Inclusion criteria: 18 y of age or older; had an occlusion of the intracranial segment of the internal carotid artery (both terminus and nonterminus occlusions) or the first or proximal second segment of the middle cerebral artery, or both; within 4.5 h after symptom onset; had a neurological deficit as indicated by a score of at least 2 points on the NIHSSIntervention: Endovascular thrombectomy alone (thrombectomy alone group; n=327). Comparator: Endovascular thrombectomy preceded by intravenous alteplase, at a dose of 0.9 mg per kilogram of body weight, administered within 4.5 h after symptom onset (combination-therapy group; n=329)Primary end point: Endovascular thrombectomy alone was noninferior to combined intravenous alteplase and endovascular thrombectomy with regard to the primary outcome (adjusted common OR 1.07 [95% CI, 0.81–1.40], P=0.04 for noninferiority) but was associated with lower percentages of patients with successful reperfusion before thrombectomy (2.4% vs 7.0%) and overall successful reperfusion (79.4% vs 84.5%). Safety end point: Similar in the two groups (37.0% [121 of 327 patients] in the thrombectomy alone group and 36.8% [121 of 329] in the combination-therapy group).
MR CLEAN–NO IV; LeCouffe et al80; 2021Study aim: to study the value of administering intravenous alteplase before EVT for acute ischemic stroke in non-Asian populations. Study type: Open-label, multicenter, randomized trial in Europe Study size: 539Inclusion criteria: 18 y of age or older; had acute ischemic stroke due to an intracranial proximal occlusion of the anterior circulation; were eligible for EVT and intravenous alteplase administration within 4.5 h after symptom onset and were admitted directly to a center that performed EVT; had a score of 2 or more on NIHSSIntervention: Endovascular thrombectomy alone (n=273). Comparator: Intravenous alteplase followed by endovascular thrombectomy (n=266)Primary end point: The median score on the mRS at 90 days was 3 (interquartile range, 2 to 5) with EVT alone and 2 (interquartile range, 2 to 5) with alteplase plus EVT. The adjusted common OR was 0.84 (95% CI, 0.62–1.15, P=0.28), which showed neither superiority nor noninferiority of EVT alone. Safety end point: Mortality was 20.5% with EVT alone and 15.8% with alteplase plus EVT (adjusted OR, 1.39 [95% CI, 0.84–2.30]). Symptomatic intracerebral hemorrhage occurred in 5.9% and 5.3% of the patients in the respective groups (adjusted OR, 1.30 [95% CI, 0.60–2.81]).
ASPECTS indicates Alberta Stroke Program Early CT Score; CT, computed tomography; CTA, CT angiography; DEVT, Direct Endovascular Thrombectomy Versus Combined IVT and Endovascular Thrombectomy for Patients With Acute Large Vessel Occlusion in the Anterior Circulation; DIRECT-MT, Direct Intraarterial Thrombectomy in order to Revascularize Acute Ischemic Stroke Patients With LVO Efficiently in Chinese Tertiary Hospitals: a Multicenter Randomized Clinical Trial; DWI, diffusion-weighted imaging; EVT, endovascular therapy; ICA, internal carotid artery; LVO, large vessel occlusion; MR CLEAN–NO IV, Multicenter Randomized Clinical Trial of Endovascular Treatment for Acute Ischemic Stroke in the Netherlands: intravenous treatment followed by intra-arterial treatment versus direct intra-arterial treatment for acute ischemic stroke caused by a proximal intracranial occlusion; MRA, magnetic resonance angiography; NIHSS, National Institutes of Health Stroke Scale; OR, odds ratio; RCT, randomized controlled trial; RR, risk ratio; and SKIP, Direct Mechanical Thrombectomy in Acute LVO Stroke.
In Europe, the MR CLEAN–NO IV trial (Multicenter Randomized Clinical Trial of Endovascular Treatment for Acute Ischemic Stroke in the Netherlands: intravenous treatment followed by intra-arterial treatment versus direct intra-arterial treatment for acute ischemic stroke caused by a proximal intracranial occlusion) included 539 patients with acute ischemic stroke due to an intracranial proximal occlusion of the anterior circulation in 20 hospitals in the Netherlands, Belgium, and France. The adjusted common OR for direct to thrombectomy versus bridging therapy was 0.84 (95% CI, 0.62–1.15]; P=0.28), which showed neither superiority nor noninferiority for direct to thrombectomy. The mortality and sICH rates were not significantly different between the two arms. However, the occlusion site was not balanced between the treatment arms, as the direct thrombectomy group had more terminal internal carotid occlusions (25% versus 18.8%, respectively), which have a worse prognosis.80 Consistent with the MR CLEAN–NO IV study, the SWIFT DIRECT trial (Solitaire With the Intention for Thrombectomy Plus Intravenous t-PA Versus DIRECT Solitaire Stent-Retriever Thrombectomy in Acute Anterior Circulation Stroke) failed to prove that direct to thrombectomy was not inferior to bridging therapy. Four hundred twenty-three patients with acute ischemic stroke due to ICA or MCA M1 occlusion in 44 academic tertiary medical centers in Europe and North America were randomly assigned to direct to thrombectomy group or bridging group. The 90-day mRS score 0 to 2 rates in the bridging group and direct to thrombectomy group were 62% and 58%, respectively, with a risk difference of −7.3% (95% CI, 17%–2.1%). The lower limit of 1-sided 95% CI was −15.1%, it did not reach the prespecified noninferiority boundary of −12%. However, the postoperative recanalization rate in the bridging group was significantly higher than that in the direct to thrombectomy group (97% versus 91%, P=0.022), and the sICH rate was also significantly higher than that in the direct to thrombectomy group (4.9% versus 1.5%, P=0.033).83 DIRECT-SAFE (A Randomized Controlled Trial of DIRECT Endovascular Clot Retrieval Versus Standard Bridging Thrombolysis With Endovascular Clot Retrieval Within 4.5 Hours of Stroke Onset; https://www.clinicaltrials.gov; Unique identifier: NCT03494920) recruited subjects from 25 subcenters in Australia, New Zealand, China, and Vietnam, with half of the enrolled subjects being East Asian and the other half was whites. The 90-day mRS score of 0 to 2 rates in the bridging group and direct to thrombectomy group was 54.8% and 60.5%, respectively, with an intention to treat analysis risk difference of −0.051 (95% CI, −0.16 to 0.059, P=0.19) after adjusting for age / NIHSS. The direct to thrombectomy group failed to meet noninferiority criteria (−0.1). The study found that bridging therapy was superior to direct to thrombectomy in East Asian subjects, but no significant differences were found between the two treatments for white subjects. This indicates that bridging therapy may be more suitable for Asians and it is the first large-scale RCT study that found that acute ischemic stroke reperfusion therapy was influenced by ethnicity and race.84
The DIRECT-MT,79 DEVT,77 and SKIP78 were challenged because of the door-to-needle time (median 59 minutes, median 61 minutes, and mean 50 minutes, respectively), which represented a substantial delay in relation to the 40 min reported in the HERMES meta-analysis (Highly Effective Reperfusion Evaluatedin Multiple Endovascular Stroke Trials)9 and the MR CLEAN–NO IV trial80 (median 31 minutes). In the DIRECT-MT study,79 the time from IVT to EVT was fast (5 minutes), about 86.5% of patients had tPA infusion during EVT procedure which may have obscured the effects of tPA in the group receiving bridge therapy. All of these 5 trials except SKIP78 were performed in patients presenting directly to a center capable of performing thrombectomy and did not include patients who were given alteplase at an outlying hospital and transferred to a larger hospital for thrombectomy. The current results do not support the discontinuation of intravenous tPA as bridging therapy, but they enhance individualization in the decision-making for LVO patients presenting directly to thrombectomy-capable centers. In this context, bridging therapy may still be advantageous in a large proportion of LVO patients. As mentioned, tenecteplase is easier and faster to administer than tPA and has higher rates of recanalization in patients with LVO who are subsequently treated with thrombectomy. Therefore, further trials are warranted to compare direct to thrombectomy with bridging therapy utilizing tenecteplase, especially trials involving drip and ship patients.
Recommendations
EVT in the anterior circulation is highly effective in the early time window after ischemic stroke onset and treatment should be initiated as soon as possible.
EVT in the anterior circulation is highly effective in patients carefully selected by advanced imaging even up to 24 hours after stroke onset.
Ongoing trials will help to define the benefits of EVT based upon baseline ischemic core size, stroke severity, and thrombus location.
EVT is of uncertain value in the posterior circulation.
Currently, it is uncertain if thrombolysis should be used before thrombectomy or if it should be skipped in certain clinical situations.

Concomitant Approaches With Thrombolysis or Thrombectomy

Although several concomitant approaches (antiplatelet therapy and anticoagulation) have been studied, evidence remains limited. A retrospective analysis of consecutive acute ischemic stroke patients admitted to a single center in Seoul, South Korea, found no increased risk of hemorrhage with early initiation of antiplatelet or anticoagulant therapy (<24 hours) after intravenous tPA or EVT compared with initiation >24 hours.85 However, this study may have had selection bias, and the timing of initiation of antiplatelet therapy or anticoagulation should be made on an individual basis, balancing risk and benefit.
There are 6 completed phase II trials suggesting that the combination therapies of argatroban plus intravenous tPA86–88 and the glycoprotein IIb/IIIa inhibitor eptifibatide plus intravenous tPA is potentially beneficial without safety concerns, supporting proceeding with phase 3 trials.89–91 The ongoing randomized trial, the MOST (Multiarm Optimization of Stroke Thrombolysis) is testing the efficacy and safety of these 2 drugs versus placebo to improve the 90-day mRS in acute ischemic stroke patients treated with thrombolysis within 3 hours of symptom onset (https://www.clinicaltrials.gov; Unique identifier: NCT03735979).

Cytoprotection

The plethora of prior therapeutic approaches targeted at the cellular consequences of focal ischemic brain injury were all unsuccessful. There are multiple reasons for these prior failures and they include both preclinical testing in animal stroke models and clinical trial design and implementation as outlined in Table 3.4 Regarding preclinical modeling deficiencies, the lessons learned from past failures have led to strengthening of the rigor of current preclinical evaluation programs of purported cytoprotective molecules as reviewed in Table 4.92 In the future, most cytoprotective drugs will be developed in conjunction with reperfusion therapy, as an adjunct, so the preclinical modeling assessment should focus on temporary occlusion models. Evaluation in permanent occlusion stroke models should also be performed, especially when the cytoprotective drug under development will be evaluated in clinical trials that focus on slowing down the evolution of the ischemic penumbra into ischemic core. For drugs that will target reperfusion injury, preclinical testing should only be done in temporary occlusion stroke models with the initiation of the drug after reperfusion to anticipate how the clinical trials will likely be conducted. A recent advance in the approach to the preclinical assessment of purported cytoprotective drugs has been the implementation of laboratory networks at multiple sites akin to what is done in later-stage clinical trials. In the United States, the National Institutes of Health–funded SPAN (Stroke Preclinical Assessment Network) which is currently evaluating five cytoprotective drugs in six laboratories that were selected by peer review.93 A similar pilot approach was done previously in Europe. The rationale for this multicenter evaluation approach is that before cytoprotective drug candidates advance to clinical trials they should demonstrate favorable outcomes in stroke models in several laboratories that perform rigorous and careful stroke modeling experiments, that is, the beneficial effects of treatment are reproducible and robust.
Table 3. Potential Reasons to Explain Why Prior Cytoprotection Trials Failed
Preclinical
 Drugs were only tested at the time of stroke onset or shortly thereafter
 Drugs were evaluated only in younger animals
 Drugs were not evaluated in animals with comorbid conditions such as hypertension or diabetes
 Drugs were exclusively evaluated in male animals and females may respond differently
 Only infarct volume was used to evaluate efficacy and behavioral or imaging end points were not assessed
 Drugs were only shown to be effective in permanent or temporary occlusion models, but not in both
 The sample sizes evaluated were inadequate and efficacy was not determined in several species in multiple laboratories
 Dose-ranging was not performed to determine the minimally effective and maximally tolerated dose
Clinical
 Drugs were evaluated too late after stroke onset
 Advanced imaging was not used to determine that substantial amounts of ischemic penumbra were still present
 For drugs only found to be effective in temporary occlusion models, randomization after i.v. thrombolysis was not required and imaging confirmation of reperfusion was not assessed.
 Only one dose was evaluated in comparison to placebo
 Only one component of the complex ischemic cascade was targeted
 The sample size was inadequate to assess a modest treatment, such as 5% absolute difference in the primary outcome measure
 Patients with lacunar stroke were included for drugs without preclinical evidence of efficacy in white matter injury
i.v indicates intravenous.
Table 4. Recommended Approaches to Improving the Preclinical Evaluation of Cytoprotective Drugs92
1. The mechanism of drug action should be established
2. Adequate dose-response and time window studies should be performed
3. Preclinical studies should be blinded, randomized, incorporate monitoring of physiological variables, have an adequate sample size, and document the adequacy of vessel occlusion
4. Outcome measures should include both histological and behavioral assessments4. Outcome measures should include both histological and behavioral assessments
5. Inclusion/exclusion criteria should be clearly stated and all animals evaluated should be accounted for, i.e.ie, the reasons for not including animals in the study outcomes should be provided
6. In later phase studies, both sexes, aged animals and animals with comorbid conditions should be evaluated
7. Positive results should be confirmed in more than one species and laboratory. Evaluation in a gyrencephalic species should be considered
8. Potential investigator conflicts of interest should be disclosed
In the past, cytoprotective drugs under development typically targeted one aspect of the ischemic cascade, and drugs that had multiple targets were considered to be flawed. With our enhanced understanding of the ischemic cascade in neurons and its complexity, as well as the roles of the other components of the neurovascular unit, drugs that affect multiple aspects of the ischemic cascade and the neurovascular unit actually appears to be more attractive because they may have a better chance to impede focal ischemic brain injury in several different ways. Previous clinical trials of potential cytoprotective drugs predominantly included patients who did not receive thrombolytic therapy so most of the patients included had an occluded blood vessel at the time the drug was initially given. With reperfusion, drug delivery will be enhanced, so in the future, if a candidate cytoprotective drug is given after imaging confirmed reperfusion, then drug delivery to the target brain tissue should be enhanced. Another potential explanation for the plethora of unsuccessful cytoprotective drug development programs is that patients were typically included in clinical trials at late time points after stroke onset.94 Advanced imaging was not used to determine if patients had persistence of a substantial amount of ischemic penumbra as a mechanism to identify patients likely to benefit, nor were slow progressors distinguished from more rapid progressors. In some prior trials, safety concerns reduced the dose that could be given and likely affected the chance of a beneficial treatment effect.
The future development of cytoprotective drugs for acute ischemic stroke in the era of highly effective reperfusion therapy can be envisioned in several ways. The first possibility is to administer the cytoprotective drug before thrombectomy. Many patients with LVO related ischemic stroke who are candidates for thrombectomy initially present at smaller hospitals where the procedure is not currently available. If their clinical and imaging evaluation at the outlying hospital identifies them as potential thrombectomy candidates, they must be transferred to a thrombectomy-capable center. This interfacility transfer can take several hours or longer depending upon the distance between the two facilities and also on traffic patterns. In patients with a substantial amount of ischemic penumbra, progression to irreversible injury occurs at variable rates and is problematic in fast progressors. A potential role for cytoprotection would be to slow down this progression during transport. A clinical trial can be envisioned using a hub and spoke model.11 Patients presenting to the smaller hospital would be enrolled there where they are initially evaluated. If they demonstrate appropriate clinical deficits and their imaging studies confirm an LVO and significant mismatch between the ischemic core and penumbra, they would then be randomized to the study cytoprotective drug versus placebo. After transport to the thrombectomy center, they would undergo repeat imaging to assess whether the ischemic core has enlarged. The primary end point of this type of phase 2 trial would be ischemic core growth on advanced imaging with the drug versus placebo. A secondary end point could be the percentage of patients in the two groups who remain thrombectomy candidates, if ongoing studies determine that thrombectomy is futile when the ischemic core exceeds a certain threshold. Typical clinical outcome data would also be collected to help inform the design and adequate sample size estimates for a phase 3 clinical efficacy trial. For such a trial, the candidate cytoprotective drug should have demonstrated a reduction of infarct size and improved functional outcome in permanent occlusion animal stroke models and ideally a beneficial effect on ischemic core growth in vivo with the use of advanced imaging techniques should also be identified.
A trial with initiation of treatment in the ambulance is another potential approach to clinical trial design before thrombectomy. The FAST-MAG trial (Field Administration of Stroke Therapy-/Magnesium) of magnesium was the first phase 3 clinical trial to demonstrate that an acute stroke trial could be performed in the prehospital setting.95 With the advent of mobile stroke unit ambulances equipped with CT scanners, appropriately trained personnel and telemedicine capability, an ambulance-based trial in thrombectomy candidates is now feasible in some locations. MSUs have demonstrated improved outcomes for onboard thrombolysis as compared to transporting patients to hospital for initiation of treatment.18,19 A trial can be designed in a similar manner to a facility to facility transfer trial with the help of a vascular neurologist at the thrombectomy-capable center via telemedicine who can evaluate the patient in conjunction with the ambulance staff and also help to interpret the head CT and CT angiogram obtained in the ambulance. Appropriate patients can then be consented and randomized while being transported to the thrombectomy center and the primary outcome would again likely be growth of ischemic core after initiation of the study drug versus placebo.
Another type of clinical trial for cytoprotection initiated before thrombectomy would be randomization of thrombectomy eligible patients at the thrombectomy-capable center before the procedure. If the study drug can be infused rapidly, its administration would be completed before thrombectomy and if a more prolonged administration is needed then it can be continued during and after the procedure. This type of trial could also include patients who are being transferred from outlying hospitals with the capability to perform the necessary imaging studies required for enrollment. Such a trial would have a clinical efficacy primary outcome measure such as the 90-day mRS that is typically used for determining efficacy in acute stroke therapy trials. Such a trial would evaluate if the cytoprotective drug conferred additional benefit to that observed with thrombectomy alone and would probably require a substantial number of patients to be adequately powered.
The recently completed phase 3 trial of Nerinetide, a PSD-95 (post-synaptic density protein-95) inhibitor, was the first such trial.96 In the ESCAPE-NA1 trial (Safety and efficacy of nerinetide [NA-1] in subjects undergoing endovascular thrombectomy for stroke) thrombectomy candidates without an extensive ischemic core and moderate to good collaterals were randomized to intravenous Nerinetide or placebo before thrombectomy at an outlying hospital or the thrombectomy-capable center. At 90 days, a favorable mRS outcome of 0 to 2 was achieved in 59.2% of the placebo group and 61.4% in the Nerinetide group, a nonstatistically significant difference. However, in a prespecified subgroup analysis of patients who did not receive tPA before randomization, a 9.5% absolute difference of favorable outcome with Nerinetide was observed that was statistically significant. This benefit of Nerinetide in patients who did not receive tPA is plausible because the plasma levels of Nerinetide were substantially lower in patients who had received tPA, and it is now known that the peptide plasmin produced by the interaction of tPA and plasminogen cleaves Nerinetide and inactivates it. In a comprehensive series of subsequent experiments, it was determined that concomitant administration of Nerinetide and tPA significantly reduced the plasma levels of Nerinetide, as in the clinical trials.97 However, if Nerinetide was given 30 minutes before tPA, no such reduction in plasma concentration was seen and a significant reduction of infarct volume was detected that was not observed when the 2 drugs were given at the same time. Another phase 3 clinical trial of Nerinetide in thrombectomy patients is currently underway in patients who have not received tPA to determine if the results observed in the initial trial can be replicated. If the trial demonstrates a significant benefit on 90-day clinical outcome, we can anticipate that Nerinetide will become the first cytoprotective drug to do so and will likely be approved by regulatory authorities.
Lastly, clinical trials with initiation of treatment after documentation of successful reperfusion can be considered because not all of the effects of the restoration of blood flow are beneficial and during the ischemic period detrimental processes may have been initiated.98 Damage to the endothelium may occur during ischemia as well as to the blood-brain barrier. These effects could enhance the risk of hemorrhage into the region that was previously ischemic. Drugs directed at ameliorating these effects could be evaluated in clinical trials where blood-brain barrier compromise has been identified.99 After reperfusion the generation of reactive oxygen molecules and activation of the immune system with the recruitment of inflammatory cells and activation of the microglia may lead to further brain tissue injury despite reperfusion.100 This can occur with the release of cytokines, chemokines, and complement. Neutrophils recruited as part of the inflammatory response release matrix metalloproteinases which can degrade the extracellular matrix and tight cellular junctions, leading to breakdown of the blood-brain barrier.101 Apoptosis or programmed cell death initiated during ischemia may also occur at late time points after reperfusion.102 All of these mechanisms of tissue injury that occur after reperfusion are potential therapeutic targets. In the past, they were of theoretical concern, but now that we have entered the era of highly effective reperfusion with thrombectomy, they may potentially affect the ultimate clinical outcome of ischemic stroke patients. In designing clinical trials targeting the various contributors to reperfusion injury, drugs should be chosen that affect one or more of the described mechanisms. Patients should be included in the trials who have substantially reperfused as documented by a perfusion imaging study after thrombectomy and the patients should also not have an extensive ischemic core because such patients are unlikely to benefit from additional therapy. Since all of the patients included in the trial will have undergone thrombectomy, the control group of thrombectomy alone will demonstrate a substantial rate of favorable 90-day outcome. Thus, to detect a significant benefit of the study drug in addition to thrombectomy will likely require a large sample size in phase 3 trials. The possibility of delivering drugs via an intraarterial catheter after thrombectomy with direct targeting of the ischemic region may help to reduce the sample size requirements as might the careful imaging-based selection of patients to include in the trial. The design and implementation of such post-thrombectomy trials will evolve as knowledge increases and lessons are learned from the initial trials.
In conclusion, despite the dismal past of cytoprotection as a therapy for acute ischemic stroke, it may have a role in the future. Many different types of clinical trials can be envisioned and some are already underway.1 Some will be successful, but many will not be successful. All of the types of trials discussed and others not yet envisioned will inform future investigators and likely lead to a future where cytoprotection serves as an important adjunctive therapy to reperfusion to maximize favorable outcomes after acute ischemic stroke.
Article Information

Footnote

Nonstandard Abbreviations and Acronyms

AcT
Alteplase Compared to Tenecteplase in Patients With Acute Ischemic Stroke
ATTEST2
Alteplase Tenecteplase Trial Evaluation for Stroke Thrombolysis
B_PROUD
Berlin_Prehospital or Usual Delivery in Stroke Care
BASICS
Basilar Artery International Cooperation Study
BEST
Basilar Artery Occlusion Endovascular Intervention Versus Standard Medical Treatment
MSU
The Benefits of Stroke Treatment Delivered by a Mobile Stroke Unit Compared with Standard Management by Emergency Medical Services
BEST
Basilar Artery Occlusion Endovascular Intervention Versus Standard Medical Treatment
CHABLIS
T Chinese Acute Tissue-Based Imaging Selection for Lysis In Stroke-Tenecteplase
CPSS
Cincinnati Prehospital Stroke Scale
CT
computed tomography
DAWN
Diffusion-Weighted Imaging or Computerized Tomography Perfusion Assessment with Clinical Mismatch in the Triage of Wake-Up and Late Presenting Strokes Undergoing Neurointervention With Trevo
DEFUSE 3
Endovascular Therapy Following Imaging Evaluation for Ischemic Stroke
DEVT
Direct Endovascular Thrombectomy Versus Combined IVT and Endovascular Thrombectomy for Patients With Acute LVO in the Anterior Circulation
DIRECT ANGIO
Effect of Direct Transfer to Angiosuite on Functional Outcome in Patient With Severe Acute Stroke Treated With Thrombectomy
DIRECT-MT
Direct Intraarterial Thrombectomy to Revascularize Acute Ischemic Stroke Patients With LVO Efficiently in Chinese Tertiary Hospitals: a Multicenter Randomized Clinical Trial
DIRECT-SAFE
A Randomized Controlled Trial of DIRECT Endovascular Clot Retrieval Versus Standard Bridging Thrombolysis With Endovascular Clot Retrieval Within 4.5 Hours of Stroke Onset
DWI
diffusion-weighted imaging
ECASS-4
European Cooperative Acute Stroke Study 4
EMS
emergency medical service
EPITHET
Echoplanar Imaging Thrombolytic Evaluation Trial
ESCAPE
Endovascular Treatment for Small Core and Anterior Circulation Proximal Occlusion With Emphasis on Minimizing CT to Recanalization Times
ETERNAL-LVO
Extending the Time Window for Tenecteplase by Effective Reperfusion in Patients With LVO
EVT
endovascular therapy
EXTEND
Extending the time for Thrombolysis in Emergency Neurological Deficits
EXTEND-IA TNK
Tenecteplase Versus Alteplase Before Thrombectomy for Ischemic Stroke
EXTEND-IA
Extending the Time for Thrombolysis in Emergency Neurological Deficits—Intra-Arterial
FLAIR
fluid-attenuated inversion recovery
FRIDA
Nonimmunogenic Recombinant Staphylokinase Versus Alteplase for Patients With Acute Ischemic Stroke 4.5 Hours After Symptom Onset in Russia
GA
general anesthesia
HERMES
Highly Effective Reperfusion Evaluated in Multiple Endovascular Stroke Trials
ICA
internal carotid artery
IVT
Intravenous thrombolysis
LVO
large vessel occlusion
MCA
middle cerebral artery
MR CLEAN
Multicenter Randomized Clinical Trial of Endovascular Treatment for Acute Ischemic Stroke in the Netherlands
MR CLEAN–NO IV
Multicenter Randomized Clinical Trial of Endovascular Treatment for Acute Ischemic Stroke in the Netherlands
MRI
magnetic resonance imaging
mRS
modified Rankin Scale
MSU
Mobile stroke unit
NIHSS
National Institutes of Health Stroke Scale
NOR-TEST
Norwegian Tenecteplase Stroke Trial
OR
odds ratio
PRISMS
The Potential of rtPA for Ischemic Strokes With Mild Symptoms
RACECAT
Direct Transfer to Endovascular Center of Acute Stroke Patients With Suspected Large Vessel Occlusion in the Catalan Territory
REVASCAT
Randomized Trial of Revascularization With Solitaire FR Device Versus Best Medical Therapy in the Treatment of Acute Stroke Due to Anterior Circulation Large Vessel Occlusion Presenting Within Eight Hours of Symptom Onset
ROSE-TNK
MRI-Guided Thrombolysis for Stroke Beyond Time Window by TNK
sICH
symptomatic intracerebral hemorrhage
SPAN
Stroke Preclinical Assessment Network
SWIFT PRIME
Solitaire With the Intention for Thrombectomy as Primary Endovascular Treatment
TEMPO-1
TNK-tPA Evaluation for Minor Ischemic Stroke With Proven Occlusion
THAWS
Thrombolysis for Acute Wake-Up and Unclear Onset Strokes With Alteplase at 0.6 mg/kg
TIMELESS
Tenecteplase in Stroke Patients Between 4.5 and 24 Hours
TNK
tenecteplase
tPA
tissue-type plasminogen activator
TWIST
Tenecteplase in Wake-Up Ischemic Stroke Trial
WE-TRUST
Workflow Optimization to Reduce Time to Endovascular Reperfusion for Ultra-fast Stroke Treatment

References

1.
Chamorro Á, Lo EH, Renú A, van Leyen K, Lyden PD. The future of neuroprotection in stroke. J Neurol Neurosurg Psychiatry. 2021;92:129–135. doi: 10.1136/jnnp-2020-324283
2.
Catanese L, Tarsia J, Fisher M. Acute ischemic stroke therapy overview. Circ Res. 2017;120:541–558. doi: 10.1161/CIRCRESAHA.116.309278
3.
Fisher M. Characterizing the target of acute stroke therapy. Stroke. 1997;28:866–872. doi: 10.1161/01.str.28.4.866
4.
Lyden PD. Cerebroprotection for acute ischemic stroke: looking ahead. Stroke. 2021;52:3033–3044. doi: 10.1161/STROKEAHA.121.032241
5.
Moskowitz MA, Lo EH, Iadecola C. The science of stroke: mechanisms in search of treatments. Neuron. 2010;67:181–198. doi: 10.1016/j.neuron.2010.07.002
6.
Ospel JM, Hill MD, Kappelhof M, Demchuk AM, Menon BK, Mayank A, Dowlatshahi D, Frei D, Rempel JL, Baxter B, et al. Which acute ischemic stroke patients are fast progressors?: Results from the ESCAPE trial control arm. Stroke. 2021;52:1847–1850. doi: 10.1161/STROKEAHA.120.032950
7.
Demeestere J, Wouters A, Christensen S, Lemmens R, Lansberg MG. Review of perfusion imaging in acute ischemic stroke: from time to tissue. Stroke. 2020;51:1017–1024. doi: 10.1161/STROKEAHA.119.028337
8.
Tsivgoulis G, Kargiotis O, Alexandrov AV. Intravenous thrombolysis for acute ischemic stroke: a bridge between two centuries. Expert Rev Neurother. 2017;17:819–837. doi: 10.1080/14737175.2017.1347039
9.
Goyal M, Menon BK, van Zwam WH, Dippel DW, Mitchell PJ, Demchuk AM, Dávalos A, Majoie CB, van der Lugt A, de Miquel MA, et al; HERMES collaborators. Endovascular thrombectomy after large-vessel ischaemic stroke: a meta-analysis of individual patient data from five randomised trials. Lancet. 2016;387:1723–1731. doi: 10.1016/S0140-6736(16)00163-X
10.
Savitz SI, Baron JC, Yenari MA, Sanossian N, Fisher M. Reconsidering neuroprotection in the reperfusion era. Stroke. 2017;48:3413–3419. doi: 10.1161/STROKEAHA.117.017283
11.
Savitz SI, Baron JC, Fisher M; STAIR X Consortium. Stroke treatment academic industry Roundtable X: brain cytoprotection therapies in the reperfusion era. Stroke. 2019;50:1026–1031. doi: 10.1161/STROKEAHA.118.023927
12.
Saver JL. Time is brain–quantified. Stroke. 2006;37:263–266. doi: 10.1161/01.STR.0000196957.55928.ab
13.
Meyran D, Cassan P, Avau B, Singletary E, Zideman DA. Stroke recognition for first aid providers: a systematic review and meta-analysis. Cureus. 2020;12:e11386. doi: 10.7759/cureus.11386
14.
Nguyen TTM, van den Wijngaard IR, Bosch J, van Belle E, van Zwet EW, Dofferhoff-Vermeulen T, Duijndam D, Koster GT, de Schryver ELLM, Kloos LMH, et al. Comparison of prehospital scales for predicting large anterior vessel occlusion in the ambulance setting. JAMA Neurol. 2021;78:157–164. doi: 10.1001/jamaneurol.2020.4418
15.
Desai SM, Leslie-Mazwi TM, Hirsch JA, Jadhav AP. Optimal transfer paradigm for emergent large vessel occlusion strokes: recognition to recanalization in the RACECAT trial. J Neurointerv Surg. 2021;13:97–99. doi: 10.1136/neurintsurg-2020-017227
16.
American Heart Association. Severity-Based stroke triage algorithm for EMS 2020. https://www.heart.org/missionlifelinestroke. Accessed Jan 15, 2022.
17.
Demaerschalk BM, Raman R, Ernstrom K, Meyer BC. Efficacy of telemedicine for stroke: pooled analysis of the Stroke Team Remote Evaluation Using a Digital Observation Camera (STRokE DOC) and STRokE DOC Arizona telestroke trials. Telemed J E Health. 2012;18:230–237. doi: 10.1089/tmj.2011.0116
18.
Ebinger M, Siegerink B, Kunz A, Wendt M, Weber JE, Schwabauer E, Geisler F, Freitag E, Lange J, Behrens J, et al; Berlin_PRehospital Or Usual Delivery in stroke care (B_PROUD) study group. Association between dispatch of mobile stroke units and functional outcomes among patients with acute ischemic stroke in Berlin. JAMA. 2021;325:454–466. doi: 10.1001/jama.2020.26345
19.
Grotta JC, Yamal JM, Parker SA, Rajan SS, Gonzales NR, Jones WJ, Alexandrov AW, Navi BB, Nour M, Spokoyny I, et al. Prospective, multicenter, controlled trial of mobile stroke units. N Engl J Med. 2021;385:971–981. doi: 10.1056/NEJMoa2103879
20.
Thomalla G, Simonsen CZ, Boutitie F, Andersen G, Berthezene Y, Cheng B, Cheripelli B, Cho TH, Fazekas F, Fiehler J, et al; WAKE-UP Investigators. MRI-Guided thrombolysis for stroke with unknown time of onset. N Engl J Med. 2018;379:611–622. doi: 10.1056/NEJMoa1804355
21.
Thomalla G, Cheng B, Ebinger M, Hao Q, Tourdias T, Wu O, Kim JS, Breuer L, Singer OC, Warach S, et al; STIR and VISTA Imaging Investigators. DWI-FLAIR mismatch for the identification of patients with acute ischaemic stroke within 4·5 h of symptom onset (PRE-FLAIR): a multicentre observational study. Lancet Neurol. 2011;10:978–986. doi: 10.1016/S1474-4422(11)70192-2
22.
Koga M, Yamamoto H, Inoue M, Asakura K, Aoki J, Hamasaki T, Kanzawa T, Kondo R, Ohtaki M, Itabashi R, et al; THAWS Trial Investigators. Thrombolysis with Alteplase at 0.6 mg/kg for stroke with unknown time of onset: a randomized controlled trial. Stroke. 2020;51:1530–1538. doi: 10.1161/STROKEAHA.119.028127
23.
Ma H, Campbell BCV, Parsons MW, Churilov L, Levi CR, Hsu C, Kleinig TJ, Wijeratne T, Curtze S, Dewey HM, et al; EXTEND Investigators. Thrombolysis guided by perfusion imaging up to 9 hours after onset of stroke. N Engl J Med. 2019;380:1795–1803. doi: 10.1056/NEJMoa1813046
24.
Ringleb P, Bendszus M, Bluhmki E, Donnan G, Eschenfelder C, Fatar M, Kessler C, Molina C, Leys D, Muddegowda G, et al; ECASS-4 study group. Extending the time window for intravenous thrombolysis in acute ischemic stroke using magnetic resonance imaging-based patient selection. Int J Stroke. 2019;14:483–490. doi: 10.1177/1747493019840938
25.
Campbell BCV, Ma H, Ringleb PA, Parsons MW, Churilov L, Bendszus M, Levi CR, Hsu C, Kleinig TJ, Fatar M, et al; EXTEND, ECASS-4, and EPITHET Investigators. Extending thrombolysis to 4·5-9 h and wake-up stroke using perfusion imaging: a systematic review and meta-analysis of individual patient data. Lancet. 2019;394:139–147. doi: 10.1016/S0140-6736(19)31053-0
26.
Thomalla G, Boutitie F, Ma H, Koga M, Ringleb P, Schwamm LH, Wu O, Bendszus M, Bladin CF, Campbell BCV, et al; Evaluation of unknown Onset Stroke thrombolysis trials (EOS) investigators. Intravenous alteplase for stroke with unknown time of onset guided by advanced imaging: systematic review and meta-analysis of individual patient data. Lancet. 2020;396:1574–1584. doi: 10.1016/S0140-6736(20)32163-2
27.
Tanswell P, Modi N, Combs D, Danays T. Pharmacokinetics and pharmacodynamics of tenecteplase in fibrinolytic therapy of acute myocardial infarction. Clin Pharmacokinet. 2002;41:1229–1245. doi: 10.2165/00003088-200241150-00001
28.
Coutts SB, Dubuc V, Mandzia J, Kenney C, Demchuk AM, Smith EE, Subramaniam S, Goyal M, Patil S, Menon BK, et al; TEMPO-1 Investigators. Tenecteplase-tissue-type plasminogen activator evaluation for minor ischemic stroke with proven occlusion. Stroke. 2015;46:769–774. doi: 10.1161/STROKEAHA.114.008504
29.
Huang X, Cheripelli BK, Lloyd SM, Kalladka D, Moreton FC, Siddiqui A, Ford I, Muir KW. Alteplase versus tenecteplase for thrombolysis after ischaemic stroke (ATTEST): a phase 2, randomised, open-label, blinded endpoint study. Lancet Neurol. 2015;14:368–376. doi: 10.1016/S1474-4422(15)70017-7
30.
Logallo N, Novotny V, Assmus J, Kvistad CE, Alteheld L, Rønning OM, Thommessen B, Amthor KF, Ihle-Hansen H, Kurz M, et al. Tenecteplase versus alteplase for management of acute ischaemic stroke (NOR-TEST): a phase 3, randomised, open-label, blinded endpoint trial. Lancet Neurol. 2017;16:781–788. doi: 10.1016/S1474-4422(17)30253-3
31.
Campbell BCV, Mitchell PJ, Churilov L, Yassi N, Kleinig TJ, Dowling RJ, Yan B, Bush SJ, Thijs V, Scroop R, et al; EXTEND-IA TNK Part 2 investigators. Effect of intravenous tenecteplase dose on cerebral reperfusion before thrombectomy in patients with large vessel occlusion ischemic stroke: the EXTEND-IA TNK Part 2 randomized clinical trial. JAMA. 2020;323:1257–1265. doi: 10.1001/jama.2020.1511
32.
Haley EC, Lyden PD, Johnston KC, Hemmen TM; TNK in Stroke Investigators. A pilot dose-escalation safety study of tenecteplase in acute ischemic stroke. Stroke. 2005;36:607–612. doi: 10.1161/01.STR.0000154872.73240.e9
33.
Parsons M, Spratt N, Bivard A, Campbell B, Chung K, Miteff F, O’Brien B, Bladin C, McElduff P, Allen C, et al. A randomized trial of tenecteplase versus alteplase for acute ischemic stroke. N Engl J Med. 2012;366:1099–1107. doi: 10.1056/NEJMoa1109842
34.
Bivard A, Huang X, Levi CR, Spratt N, Campbell BCV, Cheripelli BK, Kalladka D, Moreton FC, Ford I, Bladin CF, et al. Tenecteplase in ischemic stroke offers improved recanalization: Analysis of 2 trials. Neurology. 2017;89:62–67. doi: 10.1212/WNL.0000000000004062
35.
Bivard A, Huang X, McElduff P, Levi CR, Campbell BC, Cheripelli BK, Kalladka D, Moreton FC, Ford I, Bladin CF, et al. Impact of computed tomography perfusion imaging on the response to tenecteplase in ischemic stroke: analysis of 2 randomized controlled trials. Circulation. 2017;135:440–448. doi: 10.1161/CIRCULATIONAHA.116.022582
36.
Campbell BCV, Mitchell PJ, Churilov L, Yassi N, Kleinig TJ, Dowling RJ, Yan B, Bush SJ, Dewey HM, Thijs V, et al; EXTEND-IA TNK Investigators. Tenecteplase versus alteplase before thrombectomy for ischemic stroke. N Engl J Med. 2018;378:1573–1582. doi: 10.1056/NEJMoa1716405
37.
Katsanos AH, Safouris A, Sarraj A, Magoufis G, Leker RR, Khatri P, Cordonnier C, Leys D, Shoamanesh A, Ahmed N, et al. Intravenous thrombolysis with tenecteplase in patients with large vessel occlusions: systematic review and meta-analysis. Stroke. 2021;52:308–312. doi: 10.1161/STROKEAHA.120.030220
38.
Powers WJ, Rabinstein AA, Ackerson T, Adeoye OM, Bambakidis NC, Becker K, Biller J, Brown M, Demaerschalk BM, Hoh B, et al. Guidelines for the early management of patients with acute ischemic stroke: 2019 update to the 2018 guidelines for the early management of acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2019;50:e344–e418. doi: 10.1161/STR.0000000000000211
39.
Collen D. Staphylokinase: a potent, uniquely fibrin-selective thrombolytic agent. Nat Med. 1998;4:279–284. doi: 10.1038/nm0398-279
40.
Gusev EI, Martynov MY, Nikonov AA, Shamalov NA, Semenov MP, Gerasimets EA, Yarovaya EB, Semenov AM, Archakov AI, Markin SS; FRIDA Study Group. Non-immunogenic recombinant staphylokinase versus alteplase for patients with acute ischaemic stroke 4·5 h after symptom onset in Russia (FRIDA): a randomised, open label, multicentre, parallel-group, non-inferiority trial. Lancet Neurol. 2021;20:721–728. doi: 10.1016/S1474-4422(21)00210-6
41.
Rothwell PM, Buchan AM. A new thrombolytic drug for acute ischaemic stroke. Lancet Neurol. 2021;20:687–689. doi: 10.1016/S1474-4422(21)00256-8
42.
Khatri P, Kleindorfer DO, Devlin T, Sawyer RN, Starr M, Mejilla J, Broderick J, Chatterjee A, Jauch EC, Levine SR, et al; PRISMS Investigators. Effect of alteplase vs aspirin on functional outcome for patients with acute ischemic stroke and minor nondisabling neurologic deficits: the PRISMS randomized clinical trial. JAMA. 2018;320:156–166. doi: 10.1001/jama.2018.8496
43.
Berge E, Whiteley W, Audebert H, De Marchis GM, Fonseca AC, Padiglioni C, de la Ossa NP, Strbian D, Tsivgoulis G, Turc G. European Stroke Organisation (ESO) guidelines on intravenous thrombolysis for acute ischaemic stroke. Eur Stroke J. 2021;6:I–LXII. doi: 10.1177/2396987321989865
44.
Kleindorfer DO, Towfighi A, Chaturvedi S, Cockroft KM, Gutierrez J, Lombardi-Hill D, Kamel H, Kernan WN, Kittner SJ, Leira EC, et al. 2021 guideline for the prevention of stroke in patients with stroke and transient ischemic attack: a guideline from the American Heart Association/American Stroke Association. Stroke. 2021;52:e364–e467. doi: 10.1161/STR.0000000000000375
45.
Berkhemer OA, Fransen PS, Beumer D, van den Berg LA, Lingsma HF, Yoo AJ, Schonewille WJ, Vos JA, Nederkoorn PJ, Wermer MJ, et al; MR CLEAN Investigators. A randomized trial of intraarterial treatment for acute ischemic stroke. N Engl J Med. 2015;372:11–20. doi: 10.1056/NEJMoa1411587
46.
Goyal M, Demchuk AM, Menon BK, Eesa M, Rempel JL, Thornton J, Roy D, Jovin TG, Willinsky RA, Sapkota BL, et al; ESCAPE Trial Investigators. Randomized assessment of rapid endovascular treatment of ischemic stroke. N Engl J Med. 2015;372:1019–1030. doi: 10.1056/NEJMoa1414905
47.
Jovin TG, Chamorro A, Cobo E, de Miquel MA, Molina CA, Rovira A, San Román L, Serena J, Abilleira S, Ribó M, et al; REVASCAT Trial Investigators. Thrombectomy within 8 hours after symptom onset in ischemic stroke. N Engl J Med. 2015;372:2296–2306. doi: 10.1056/NEJMoa1503780
48.
Saver JL, Goyal M, Bonafe A, Diener HC, Levy EI, Pereira VM, Albers GW, Cognard C, Cohen DJ, Hacke W, et al; SWIFT PRIME Investigators. Stent-retriever thrombectomy after intravenous t-PA vs. t-PA alone in stroke. N Engl J Med. 2015;372:2285–2295. doi: 10.1056/NEJMoa1415061
49.
Campbell BC, Mitchell PJ, Kleinig TJ, Dewey HM, Churilov L, Yassi N, Yan B, Dowling RJ, Parsons MW, Oxley TJ, et al; EXTEND-IA Investigators. Endovascular therapy for ischemic stroke with perfusion-imaging selection. N Engl J Med. 2015;372:1009–1018. doi: 10.1056/NEJMoa1414792
50.
Nogueira RG, Jadhav AP, Haussen DC, Bonafe A, Budzik RF, Bhuva P, Yavagal DR, Ribo M, Cognard C, Hanel RA, et al; DAWN Trial Investigators. Thrombectomy 6 to 24 hours after stroke with a mismatch between deficit and infarct. N Engl J Med. 2018;378:11–21. doi: 10.1056/NEJMoa1706442
51.
Albers GW, Marks MP, Kemp S, Christensen S, Tsai JP, Ortega-Gutierrez S, McTaggart RA, Torbey MT, Kim-Tenser M, Leslie-Mazwi T, et al; DEFUSE 3 Investigators. Thrombectomy for stroke at 6 to 16 hours with selection by perfusion imaging. N Engl J Med. 2018;378:708–718. doi: 10.1056/NEJMoa1713973
52.
Jovin TG, Nogueira RG, Lansberg MG, Demchuk AM, Martins SO, Mocco J, Ribo M, Jadhav AP, Ortega-Gutierrez S, Hill MD, et al. Thrombectomy for anterior circulation stroke beyond 6 h from time last known well (AURORA): a systematic review and individual patient data meta-analysis. Lancet. 2022;399:249–258. doi: 10.1016/S0140-6736(21)01341-6
53.
Albers GW, Lansberg MG, Brown S, Jadhav AP, Haussen DC, Martins SO, Rebello LC, Demchuk AM, Goyal M, Ribo M, et al; AURORA Investigators. Assessment of optimal patient selection for endovascular thrombectomy beyond 6 hours after symptom onset: a pooled analysis of the AURORA database. JAMA Neurol. 2021;78:1064–1071. doi: 10.1001/jamaneurol.2021.2319
54.
Nguyen TN, Abdalkader M, Nagel S, Qureshi MM, Ribo M, Caparros F, Haussen DC, Mohammaden MH, Sheth SA, Ortega-Gutierrez S, et al. Noncontrast computed tomography vs computed tomography perfusion or magnetic resonance imaging selection in late presentation of stroke with large-vessel occlusion. JAMA Neurol. 2022;79:22–31. doi: 10.1001/jamaneurol.2021.4082
55.
Riou-Comte N, Zhu F, Cherifi A, Richard S, Nace L, Audibert G, Achit H, Costalat V, Arquizan C, Beaufils O, et al; DIRECT ANGIO Investigators. 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. doi: 10.1136/bmjopen-2020-040522
56.
Ribo M, Boned S, Rubiera M, Tomasello A, Coscojuela P, Hernández D, Pagola J, Juega J, Rodriguez N, Muchada M, et al. Direct transfer to angiosuite to reduce door-to-puncture time in thrombectomy for acute stroke. J Neurointerv Surg. 2018;10:221–224. doi: 10.1136/neurintsurg-2017-013038
57.
Pfaff JAR, Schönenberger S, Herweh C, Ulfert C, Nagel S, Ringleb PA, Bendszus M, Möhlenbruch MA. Direct transfer to angio-suite versus computed tomography-transit in patients receiving mechanical thrombectomy: a randomized trial. Stroke. 2020;51:2630–2638. doi: 10.1161/STROKEAHA.120.029905
58.
Sarraj A, Goyal N, Chen M, Grotta JC, Blackburn S, Requena M, Kamal H, Abraham MG, Elijovich L, Dannenbaum M, et al. Direct to angiography vs repeated imaging approaches in transferred patients undergoing endovascular thrombectomy. JAMA Neurol. 2021;78:916–926. doi: 10.1001/jamaneurol.2021.1707
59.
Lapergue B, Blanc R, Gory B, Labreuche J, Duhamel A, Marnat G, Saleme S, Costalat V, Bracard S, Desal H, et al; ASTER Trial Investigators. Effect of endovascular contact aspiration vs stent retriever on revascularization in patients with acute ischemic stroke and large vessel occlusion: the ASTER randomized clinical trial. JAMA. 2017;318:443–452. doi: 10.1001/jama.2017.9644
60.
Turk AS, Siddiqui A, Fifi JT, De Leacy RA, Fiorella DJ, Gu E, Levy EI, Snyder KV, Hanel RA, Aghaebrahim A, et al. Aspiration thrombectomy versus stent retriever thrombectomy as first-line approach for large vessel occlusion (COMPASS): a multicentre, randomised, open label, blinded outcome, non-inferiority trial. Lancet. 2019;393:998–1008. doi: 10.1016/S0140-6736(19)30297-1
61.
Zaidat OO, Castonguay AC, Linfante I, Gupta R, Martin CO, Holloway WE, Mueller-Kronast N, English JD, Dabus G, Malisch TW, et al. First pass effect: a new measure for stroke thrombectomy devices. Stroke. 2018;49:660–666. doi: 10.1161/STROKEAHA.117.020315
62.
Mokin M, Waqas M, Fifi J, De Leacy R, Fiorella D, Levy EI, Snyder K, Hanel R, Woodward K, Chaudry I, et al. Clot perviousness is associated with first pass success of aspiration thrombectomy in the COMPASS trial. J Neurointerv Surg. 2021;13:509–514. doi: 10.1136/neurintsurg-2020-016434
63.
Liu Y, Brinjikji W, Abbasi M, Dai D, Arturo Larco JL, Madhani SI, Shahid AH, Mereuta OM, Nogueira RG, Kvamme P, et al. Quantification of clot spatial heterogeneity and its impact on thrombectomy. J Neurointerv Surg. 2021;13. neurintsurg-2021-018183. doi: 10.1136/neurintsurg-2021-018183
64.
Nguyen TN, Malisch T, Castonguay AC, Gupta R, Sun CH, Martin CO, Holloway WE, Mueller-Kronast N, English JD, Linfante I, et al. Balloon guide catheter improves revascularization and clinical outcomes with the Solitaire device: analysis of the North American Solitaire Acute Stroke Registry. Stroke. 2014;45:141–145. doi: 10.1161/STROKEAHA.113.002407
65.
Zaidat OO, Mueller-Kronast NH, Hassan AE, Haussen DC, Jadhav AP, Froehler MT, Jahan R, Ali Aziz-Sultan M, Klucznik RP, Saver JL, et al; STRATIS Investigators. Impact of balloon guide catheter use on clinical and angiographic outcomes in the STRATIS stroke thrombectomy registry. Stroke. 2019;50:697–704. doi: 10.1161/STROKEAHA.118.021126
66.
Coutinho JM, Liebeskind DS, Slater LA, Nogueira RG, Baxter BW, Levy EI, Siddiqui AH, Goyal M, Zaidat OO, Davalos A, et al. Mechanical thrombectomy for isolated M2 occlusions: a post hoc analysis of the STAR, SWIFT, and SWIFT PRIME studies. AJNR Am J Neuroradiol. 2016;37:667–672. doi: 10.3174/ajnr.A4591
67.
Mokin M, Ansari SA, McTaggart RA, Bulsara KR, Goyal M, Chen M, Fraser JF; Society of NeuroInterventional Surgery. Indications for thrombectomy in acute ischemic stroke from emergent large vessel occlusion (ELVO): report of the SNIS Standards and Guidelines Committee. J Neurointerv Surg. 2019;11:215–220. doi: 10.1136/neurintsurg-2018-014640
68.
Goyal N, Tsivgoulis G, Malhotra K, Ishfaq MF, Pandhi A, Frohler MT, Spiotta AM, Anadani M, Psychogios M, Maus V, et al. Medical management vs mechanical thrombectomy for mild strokes: an international multicenter study and systematic review and meta-analysis. JAMA Neurol. 2020;77:16–24. doi: 10.1001/jamaneurol.2019.3112
69.
Schönenberger S, Uhlmann L, Hacke W, Schieber S, Mundiyanapurath S, Purrucker JC, Nagel S, Klose C, Pfaff J, Bendszus M, et al. Effect of conscious sedation vs general anesthesia on early neurological improvement among patients with ischemic stroke undergoing endovascular thrombectomy: a randomized clinical trial. JAMA. 2016;316:1986–1996. doi: 10.1001/jama.2016.16623
70.
Schönenberger S, Hendén PL, Simonsen CZ, Uhlmann L, Klose C, Pfaff JAR, Yoo AJ, Sørensen LH, Ringleb PA, Wick W, et al. Association of general anesthesia vs procedural sedation with functional outcome among patients with acute ischemic stroke undergoing thrombectomy: a systematic review and meta-analysis. JAMA. 2019;322:1283–1293. doi: 10.1001/jama.2019.11455
71.
Simonsen CZ, Yoo AJ, Sørensen LH, Juul N, Johnsen SP, Andersen G, Rasmussen M. Effect of general anesthesia and conscious sedation during endovascular therapy on infarct growth and clinical outcomes in acute ischemic stroke: a randomized clinical trial. JAMA Neurol. 2018;75:470–477. doi: 10.1001/jamaneurol.2017.4474
72.
Liu X, Dai Q, Ye R, Zi W, Liu Y, Wang H, Zhu W, Ma M, Yin Q, Li M, et al; BEST Trial Investigators. Endovascular treatment versus standard medical treatment for vertebrobasilar artery occlusion (BEST): an open-label, randomised controlled trial. Lancet Neurol. 2020;19:115–122. doi: 10.1016/S1474-4422(19)30395-3
73.
Langezaal LCM, van der Hoeven EJRJ, Mont’Alverne FJA, de Carvalho JJF, Lima FO, Dippel DWJ, van der Lugt A, Lo RTH, Boiten J, Lycklama À Nijeholt GJ, et al; BASICS Study Group. Endovascular therapy for stroke due to basilar-artery occlusion. N Engl J Med. 2021;384:1910–1920. doi: 10.1056/NEJMoa2030297
74.
Fisher M. Endovascular therapy for basilar-artery occlusion - still waiting for answers. N Engl J Med. 2021;384:1954–1955. doi: 10.1056/NEJMe2104814
75.
Alemseged F, Van der Hoeven E, Di Giuliano F, Shah D, Sallustio F, Arba F, Kleinig TJ, Bush S, Dowling RJ, Yan B, et al. Response to late-window endovascular revascularization is associated with collateral status in basilar artery occlusion. Stroke. 2019;50:STROKEAHA118023361. doi: 10.1161/STROKEAHA.118.023361
76.
Chandra RV, Leslie-Mazwi TM, Mehta BP, Derdeyn CP, Demchuk AM, Menon BK, Goyal M, González RG, Hirsch JA. Does the use of IV tPA in the current era of rapid and predictable recanalization by mechanical embolectomy represent good value? J Neurointerv Surg. 2016;8:443–446. doi: 10.1136/neurintsurg-2015-012231
77.
Zi W, Qiu Z, Li F, Sang H, Wu D, Luo W, Liu S, Yuan J, Song J, Shi Z, et al; DEVT Trial Investigators. Effect of endovascular treatment alone vs intravenous alteplase plus endovascular treatment on functional independence in patients with acute ischemic stroke: the DEVT randomized clinical trial. JAMA. 2021;325:234–243. doi: 10.1001/jama.2020.23523
78.
Suzuki K, Matsumaru Y, Takeuchi M, Morimoto M, Kanazawa R, Takayama Y, Kamiya Y, Shigeta K, Okubo S, Hayakawa M, et al; SKIP Study Investigators. Effect of mechanical thrombectomy without vs with intravenous thrombolysis on functional outcome among patients with acute ischemic stroke: the SKIP randomized clinical trial. JAMA. 2021;325:244–253. doi: 10.1001/jama.2020.23522
79.
Yang P, Zhang Y, Zhang L, Zhang Y, Treurniet KM, Chen W, Peng Y, Han H, Wang J, Wang S, et al; DIRECT-MT Investigators. Endovascular thrombectomy with or without intravenous alteplase in acute stroke. N Engl J Med. 2020;382:1981–1993. doi: 10.1056/NEJMoa2001123
80.
LeCouffe NE, Kappelhof M, Treurniet KM, Rinkel LA, Bruggeman AE, Berkhemer OA, Wolff L, van Voorst H, Tolhuisen ML, Dippel DWJ, et al; MR CLEAN–NO IV Investigators. A randomized trial of intravenous alteplase before endovascular treatment for stroke. N Engl J Med. 2021;385:1833–1844. doi: 10.1056/NEJMoa2107727
81.
Lin CJ, Saver JL. Noninferiority margins in trials of thrombectomy devices for acute ischemic stroke: is the bar being set too low? Stroke. 2019;50:3519–3526. doi: 10.1161/STROKEAHA.119.026717
82.
Katsanos AH, Turc G, Psychogios M, Kaesmacher J, Palaiodimou L, Stefanou MI, Magoufis G, Shoamanesh A, Themistocleous M, Sacco S, et al. Utility of intravenous alteplase prior to endovascular stroke treatment: a systematic review and meta-analysis of RCTs. Neurology. 2021;97:e777–e784. doi: 10.1212/WNL.0000000000012390
83.
Gralla J, Fischer U. Solitaire™ with the intention for thrombectomy plus intravenous t-pa versus direct solitaire™ stent-retriever thrombectomy in acute anterior circulation stroke (SWIFT DIRECT): Abstract. Presented at 7th European Stroke Organisation Conference (ESOC 2021; 1–3 September, virtual).
84.
Yan B. A Randomized Controlled Trial of DIRECT Endovascular Clot Retrieval Versus Standard Bridging Thrombolysis With Endovascular Clot Retrieval Within 4.5 Hours of Stroke Onset (DIRECT-SAFE): Abstract. Presented at 13th World Stroke Congress (WSC 2021; 28–29 October, virtual).
85.
Jeong HG, Kim BJ, Yang MH, Han MK, Bae HJ, Lee SH. Stroke outcomes with use of antithrombotics within 24 hours after recanalization treatment. Neurology. 2016;87:996–1002. doi: 10.1212/WNL.0000000000003083
86.
Berekashvili K, Soomro J, Shen L, Misra V, Chen PR, Blackburn S, Dannenbaum M, Grotta JC, Barreto AD. Safety and feasibility of Argatroban, Recombinant Tissue Plasminogen Activator, and Intra-Arterial Therapy in Stroke (ARTSS-IA Study). J Stroke Cerebrovasc Dis. 2018;27:3647–3651. doi: 10.1016/j.jstrokecerebrovasdis.2018.08.036
87.
Barreto AD, Ford GA, Shen L, Pedroza C, Tyson J, Cai C, Rahbar MH, Grotta JC; ARTSS-2 Investigators. Randomized, multicenter Trial of ARTSS-2 (Argatroban With Recombinant Tissue Plasminogen Activator for Acute Stroke). Stroke. 2017;48:1608–1616. doi: 10.1161/STROKEAHA.117.016720
88.
Barreto AD, Alexandrov AV, Lyden P, Lee J, Martin-Schild S, Shen L, Wu TC, Sisson A, Pandurengan R, Chen Z, et al. The argatroban and tissue-type plasminogen activator stroke study: final results of a pilot safety study. Stroke. 2012;43:770–775. doi: 10.1161/STROKEAHA.111.625574
89.
Pancioli AM, Broderick J, Brott T, Tomsick T, Khoury J, Bean J, del Zoppo G, Kleindorfer D, Woo D, Khatri P, et al; CLEAR Trial Investigators. The combined approach to lysis utilizing eptifibatide and rt-PA in acute ischemic stroke: the CLEAR stroke trial. Stroke. 2008;39:3268–3276. doi: 10.1161/STROKEAHA.108.517656
90.
Pancioli AM, Adeoye O, Schmit PA, Khoury J, Levine SR, Tomsick TA, Sucharew H, Brooks CE, Crocco TJ, Gutmann L, et al; CLEAR-ER Investigators. Combined approach to lysis utilizing eptifibatide and recombinant tissue plasminogen activator in acute ischemic stroke-enhanced regimen stroke trial. Stroke. 2013;44:2381–2387. doi: 10.1161/STROKEAHA.113.001059
91.
Adeoye O, Sucharew H, Khoury J, Vagal A, Schmit PA, Ewing I, Levine SR, Demel S, Eckerle B, Katz B, et al. Combined approach to lysis utilizing eptifibatide and recombinant tissue-type plasminogen activator in acute ischemic stroke-full dose regimen stroke trial. Stroke. 2015;46:2529–2533. doi: 10.1161/STROKEAHA.115.010260
92.
Lyden P, Buchan A, Boltze J, Fisher M; STAIR XI Consortium*. Top priorities for cerebroprotective studies-a paradigm shift: report From STAIR XI. Stroke. 2021;52:3063–3071. doi: 10.1161/STROKEAHA.121.034947
93.
Dirnagl U, Hakim A, Macleod M, Fisher M, Howells D, Alan SM, Steinberg G, Planas A, Boltze J, Savitz S, et al. A concerted appeal for international cooperation in preclinical stroke research. Stroke. 2013;44:1754–1760. doi: 10.1161/STROKEAHA.113.000734
94.
Henninger N, Kumar R, Fisher M. Acute ischemic stroke therapy. Expert Rev Cardiovasc Ther. 2010;8:1389–1398. doi: 10.1586/erc.10.128
95.
Saver JL, Starkman S, Eckstein M, Stratton SJ, Pratt FD, Hamilton S, Conwit R, Liebeskind DS, Sung G, Kramer I, et al; FAST-MAG Investigators and Coordinators. Prehospital use of magnesium sulfate as neuroprotection in acute stroke. N Engl J Med. 2015;372:528–536. doi: 10.1056/NEJMoa1408827
96.
Hill MD, Goyal M, Menon BK, Nogueira RG, McTaggart RA, Demchuk AM, Poppe AY, Buck BH, Field TS, Dowlatshahi D, et al; ESCAPE-NA1 Investigators. 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. doi: 10.1016/S0140-6736(20)30258-0
97.
Mayor-Nunez D, Ji Z, Sun X, Teves L, Garman JD, Tymianski M. Plasmin-resistant PSD-95 inhibitors resolve effect-modifying drug-drug interactions between alteplase and nerinetide in acute stroke. Sci Transl Med. 2021;13:eabb1498. doi: 10.1126/scitranslmed.abb1498
98.
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. doi: 10.1111/ijs.12434
99.
Luby M, Hsia AW, Nadareishvili Z, Cullison K, Pednekar N, Adil MM, Latour LL. Frequency of blood-brain barrier disruption post-endovascular therapy and multiple thrombectomy passes in acute ischemic stroke patients. Stroke. 2019;50:2241–2244. doi: 10.1161/STROKEAHA.119.025914
100.
Mizuma A, Yenari MA. Anti-Inflammatory targets for the treatment of reperfusion injury in stroke. Front Neurol. 2017;8:467. doi: 10.3389/fneur.2017.00467
101.
Yang C, Hawkins KE, Doré S, Candelario-Jalil E. Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am J Physiol Cell Physiol. 2019;316:C135–C153. doi: 10.1152/ajpcell.00136.2018
102.
Radak D, Katsiki N, Resanovic I, Jovanovic A, Sudar-Milovanovic E, Zafirovic S, Mousad SA, Isenovic ER. Apoptosis and acute brain ischemia in ischemic stroke. Curr Vasc Pharmacol. 2017;15:115–122. doi: 10.2174/1570161115666161104095522

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Circulation Research
Go to Circulation Research
Circulation Research
Pages: 1230 - 1251
PubMed: 35420919

History

Published online: 14 April 2022
Published in print: 15 April 2022

Permissions

Request permissions for this article.

Keywords

  1. cytoprotection
  2. ischemic stroke
  3. reperfusion
  4. tenecteplase
  5. thrombectomy

Subjects

Authors

Affiliations

Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (Y.X.).
Chinese Institute of Brain Research (Y.X.).
Department of Neurointerventional Radiology Beth Israel Lahey Health Medical Center, Tufts University School of Medicine, Burlington‚ MA (A.K.W.).
Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School‚ Boston‚ MA (M.F.).

Notes

For Disclosures, see page 1248.
Correspondence to: Marc Fisher, MD, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215. Email [email protected]

Disclosures

Disclosures None.

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. A novel framework for uncovering the coordinative spectrum-effect correlation of the effective components of Yangyin Tongnao Granules on cerebral ischemia-reperfusion injury in rats, Journal of Ethnopharmacology, 337, (118844), (2025).https://doi.org/10.1016/j.jep.2024.118844
    Crossref
  2. Sex Disparities in the Direct Cost and Management of Stroke: A Population-Based Retrospective Study, Healthcare, 12, 14, (1369), (2024).https://doi.org/10.3390/healthcare12141369
    Crossref
  3. Pre-Stroke Antihypertensive Therapy Affects Stroke Severity and 3-Month Outcome of Ischemic MCA-Territory Stroke, Diseases, 12, 3, (53), (2024).https://doi.org/10.3390/diseases12030053
    Crossref
  4. Human-Induced Pluripotent Stem Cell-Derived Neural Progenitor Cells Showed Neuronal Differentiation, Neurite Extension, and Formation of Synaptic Structures in Rodent Ischemic Stroke Brains, Cells, 13, 8, (671), (2024).https://doi.org/10.3390/cells13080671
    Crossref
  5. Blocking of microglia-astrocyte proinflammatory signaling is beneficial following stroke, Frontiers in Molecular Neuroscience, 16, (2024).https://doi.org/10.3389/fnmol.2023.1305949
    Crossref
  6. A novel nomogram to predict futile recanalization in patients with acute ischemic stroke undergoing mechanical thrombectomy, Frontiers in Neurology, 15, (2024).https://doi.org/10.3389/fneur.2024.1367950
    Crossref
  7. Harnessing filamentous phages for enhanced stroke recovery, Frontiers in Immunology, 14, (2024).https://doi.org/10.3389/fimmu.2023.1343788
    Crossref
  8. Causal relationship between novel antidiabetic drugs and ischemic stroke: a drug-targeted Mendelian randomization study, Frontiers in Cardiovascular Medicine, 11, (2024).https://doi.org/10.3389/fcvm.2024.1449185
    Crossref
  9. Application of stimuli-responsive nanomedicines for the treatment of ischemic stroke, Frontiers in Bioengineering and Biotechnology, 11, (2024).https://doi.org/10.3389/fbioe.2023.1329959
    Crossref
  10. Internet + wearable device training effects on limb function recovery and serum neurocytokine content in stroke patients, NeuroRehabilitation, 55, 1, (17-28), (2024).https://doi.org/10.3233/NRE-230347
    Crossref
  11. See more
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

Advances in Acute Ischemic Stroke Therapy
Circulation Research
  • Vol. 130
  • No. 8

Purchase access to this journal for 24 hours

Circulation Research
  • Vol. 130
  • No. 8
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Media

Figures

Other

Tables

Share

Share

Share article link

Share

Comment Response