Safety and Efficacy of Otaplimastat in Patients with Acute Ischemic Stroke Requiring tPA (SAFE-TPA): A Multicenter, Randomized, Double-Blind, Placebo-Controlled Phase 2 Study

Jong S Kim, Kyung Bok Lee, Jong-Ho Park, Sang Min Sung, Kyungmi Oh, Eung-Gyu Kim, Dae-Il Chang, Yang Ha Hwang, Eun-Jae Lee, Won-Ki Kim, Chung Ju, Byung Su Kim, Jei-Man Ryu, SAFE-TPA Investigators, Jong S Kim, Kyung Bok Lee, Jong-Ho Park, Sang Min Sung, Kyungmi Oh, Eung-Gyu Kim, Dae-Il Chang, Yang Ha Hwang, Eun-Jae Lee, Won-Ki Kim, Chung Ju, Byung Su Kim, Jei-Man Ryu, SAFE-TPA Investigators

Abstract

Objective: Otaplimastat is a neuroprotectant that inhibits matrix metalloprotease pathway, and reduces edema and intracerebral hemorrhage induced by recombinant tissue plasminogen activator (rtPA) in animal stroke models. We aimed to assess the safety and efficacy of otaplimastat in patients receiving rtPA.

Methods: This was a phase 2, 2-part, multicenter trial in stroke patients (19-80 years old) receiving rtPA. Intravenous otaplimastat was administered <30 minutes after rtPA. Stage 1 was a single-arm, open-label safety study in 11 patients. Otaplimastat 80 mg was administered twice daily for 3 days. Stage 2 was a randomized, double-blind, placebo-controlled study involving 69 patients, assigned (1:1:1) to otaplimastat 40 mg, otaplimastat 80 mg, or a placebo. The primary endpoint was the occurrence of parenchymal hematoma (PH) on day 1. Secondary endpoints included serious adverse events (SAEs), mortality, and modified Rankin scale (mRS) distribution at 90 days (clinicaltrials.gov identifier: NCT02787278).

Results: No safety issues were encountered in stage 1. The incidence of PH during stage 2 was comparable: 0 of 22 with the placebo, 0 of 22 with otaplimastat 40 mg, and 1 of 21 with the 80 mg dose. No differences in SAEs (13%, 17%, 14%) or death (8.3%, 4.2%, 4.8%) were observed among the 3 groups. Three adverse events (chills, muscle rigidity, hepatotoxicity) were judged to be related to otaplimastat.

Interpretation: Intravenous otaplimastat adjunctive therapy in patients receiving rtPA is feasible and generally safe. The functional efficacy of otaplimastat needs to be investigated with further large trials. ANN NEUROL 2020;87:233-245.

Conflict of interest statement

Nothing to report.

© 2019 American Neurological Association.

Figures

Figure 1
Figure 1
Trial profile. In stage 1, 1 patient was excluded because of low (10 g/dl) hemoglobin (exclusion criterion). In stage 2, 1 patient in the otaplimastat 40 mg group received warfarin (international normalized ratio

Figure 2

Secondary efficacy outcomes. (A) Distribution…

Figure 2

Secondary efficacy outcomes. (A) Distribution of the modified Rankin score (mRS) at 90…

Figure 2
Secondary efficacy outcomes. (A) Distribution of the modified Rankin score (mRS) at 90 days and (B) National Institutes of Health Stroke Scale (NIHSS) score changes from baseline (modified intention‐to‐treat population). mRS distribution at 90 days uses imputed data only for death cases (mRS = 6). Error bars indicate standard deviation values. mRS on day 90: placebo, n = 21; otaplimastat 40 mg, n = 20; 80 mg, n = 20. NIHSS: placebo, n = 22; otaplimastat 40 mg, n = 22; 80 mg, n = 21 on day 0 to day 2. Placebo, n = 20; otaplimastat 40 mg, n = 22; 80 mg, n = 19 on day 3. Placebo, n = 20; otaplimastat 40 mg, n = 20; 80 mg, n = 19 on days 4, 5, and 28. Placebo, n = 19; otaplimastat 40 mg, n = 17; 80 mg, n = 17 on day 90. (C) Fold change of infarct growth was calculated as (individual infarct volume on day 5 − individual infarct volume on day 0)/mean of infarct volumes on day 0. Each point represents an individual patient, with the median (red bar) and interquartile ranges (black bars). Placebo, n = 20; otaplimastat 40 mg, n = 20; 80 mg, n = 19.
Figure 2
Figure 2
Secondary efficacy outcomes. (A) Distribution of the modified Rankin score (mRS) at 90 days and (B) National Institutes of Health Stroke Scale (NIHSS) score changes from baseline (modified intention‐to‐treat population). mRS distribution at 90 days uses imputed data only for death cases (mRS = 6). Error bars indicate standard deviation values. mRS on day 90: placebo, n = 21; otaplimastat 40 mg, n = 20; 80 mg, n = 20. NIHSS: placebo, n = 22; otaplimastat 40 mg, n = 22; 80 mg, n = 21 on day 0 to day 2. Placebo, n = 20; otaplimastat 40 mg, n = 22; 80 mg, n = 19 on day 3. Placebo, n = 20; otaplimastat 40 mg, n = 20; 80 mg, n = 19 on days 4, 5, and 28. Placebo, n = 19; otaplimastat 40 mg, n = 17; 80 mg, n = 17 on day 90. (C) Fold change of infarct growth was calculated as (individual infarct volume on day 5 − individual infarct volume on day 0)/mean of infarct volumes on day 0. Each point represents an individual patient, with the median (red bar) and interquartile ranges (black bars). Placebo, n = 20; otaplimastat 40 mg, n = 20; 80 mg, n = 19.

References

    1. National Institute of Neurological Disorders and Stroke rt‐PA Stroke Study Group . Tissue plasminogen activator for acute ischemic stroke. N Engl J Med 1995;333:1581−1587.
    1. Hacke W, Kaste M, Bluhmki E, et al. Thrombolysis with alteplase 3 to 4.5 hours after acute ischemic stroke. N Engl J Med 2008;359:1317−1329.
    1. Emberson J, Lees KR, Lyden P, et al. Effect of treatment delay, age, and stroke severity on the effects of intravenous thrombolysis with alteplase for acute ischaemic stroke: a meta‐analysis of individual patient data from randomised trials. Lancet 2014;384:1929−1935.
    1. Thomalla G, Simonsen CZ, Boutitie F, et al. WAKE‐UP Investigators. MRI‐guided thrombolysis for stroke with unknown time of onset. N Engl J Med 2018;379:611−622.
    1. Wang X, Tsuji K, Lee SR, et al. Mechanisms of hemorrhagic transformation after tissue plasminogen activator reperfusion therapy for ischemic stroke. Stroke 2004;35:2726−2730.
    1. Lenglet S, Montecucco F, Mach F, et al. Analysis of the expression of nine secreted matrix metalloproteinases and their endogenous inhibitors in the brain of mice subjected to ischaemic stroke. Thromb Haemost 2014;112:363−378.
    1. Jickling GC, Liu D, Stamova B, et al. Hemorrhagic transformation after ischemic stroke in animals and humans. J Cereb Blood Flow Metab 2014;34:185−199.
    1. Chamorro A, Dirnagl U, Urra X, Planas AM. Neuroprotection in acute stroke: targeting excitotoxicity, oxidative and nitrosative stress, and inflammation. Lancet Neurol 2016;15:869−881.
    1. Pena ID, Borlongan C, Shen G, Davis W. Strategies to extend thrombolytic time window for ischemic stroke treatment: an unmet clinical need. J Stroke 2017;19:50−60.
    1. Kanazawa M, Takahashi T, Nishizawa M, Shimohata T. Therapeutic strategies to attenuate hemorrhagic transformation after tissue plasminogen activator treatment for acute ischemic stroke. J Atheroscler Thromb 2017;24:240−253.
    1. Chamorro A. Neuroprotectants in the era of reperfusion therapy. J Stroke 2018;20:197−207.
    1. Malhotra K, Chang JJ, Khunger A, et al. Minocycline for acute stroke treatment: a systematic review and meta‐analysis of randomized clinical trials. J Neurol 2018;265:1871−1879.
    1. Chamorro A, Amaro S, Castellanos M, et al. Safety and efficacy of uric acid in patients with acute stroke (URICO‐ICTUS): a randomised, double‐blind phase 2b/3 trial. Lancet Neurol 2014;13:453−460.
    1. Lyden P, Pryor KE, Coffey CS, et al. Final results of the RHAPSODY trial: a multi‐center, phase 2 trial using a continual reassessment method to determine the safety and tolerability of 3K3A‐APC, a recombinant variant of human activated protein C, in combination with tissue plasminogen activator, mechanical thrombectomy or both in moderate to severe acute ischemic stroke. Ann Neurol 2019;85:125−136.
    1. Zhu Z, Fu Y, Tian D, et al. Combination of the immune modulator fingolimod with alteplase in acute ischemic stroke: a pilot trial. Circulation 2015;132:1104−1112.
    1. Tian DC, Shi K, Zhu Z, et al. Fingolimod enhances the efficacy of delayed alteplase administration in acute ischemic stroke by promoting anterograde reperfusion and retrograde collateral flow. Ann Neurol 2018;84:717−728.
    1. Noh SJ, Lee JM, Lee KS, et al. SP‐8203 shows neuroprotective effects and improves cognitive impairment in ischemic brain injury through NMDA receptor. Pharmacol Biochem Behav 2011;100:73−80.
    1. Noh SJ, Lee SH, Shin KY, et al. SP‐8203 reduces oxidative stress via SOD activity and behavioral deficit in cerebral ischemia. Pharmacol Biochem Behav 2011;98:150−154.
    1. Kim W‐K, Ju C, Jalin AMAA, et al. Therapeutic efficacy and pharmacological mechanisms of SP‐8203 for treatment of cerebral ischemia. Stroke 2015;46:ATP239 (Abstract).
    1. Ju C, Anthony Jalin A, Song HY, et al. Extension of therapeutic time window of tissue plasminogen activator with SP‐8203 combination therapy in rat embolic stroke models. Eur Stroke J 2016;1:581 (Abstract).
    1. National Institute of Neurological Disorders and Stroke rt‐PA Stroke Study Group . Intracerebral hemorrhage after intravenous t‐PA therapy for ischemic stroke. Stroke 1997;28:2109−2118.
    1. Hacke W, Kaste M, Fieschi C, et al. Intravenous thrombolysis with recombinant tissue plasminogen activator for acute hemispheric stroke. The European Cooperative Acute Stroke Study (ECASS). JAMA 1995;274:1017−1025.
    1. Hacke W, Kaste M, Fieschi C, et al. Randomised double‐blind placebo‐controlled trial of thrombolytic therapy with intravenous alteplase in acute ischaemic stroke (ECASS II). Second European‐Australasian Acute Stroke Study Investigators. Lancet 1998;352:1245−1251.
    1. Schulman S, Kearon C, Subcommittee on Control of Anticoagulation of the Scientific Standardization Committee of the International Society on Thrombosis and Haemostasis . Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non‐surgical patients. J Thromb Haemost 2005;3:692−694.
    1. Saposnik G, Fang J, Kapral MK, et al. The iScore predicts effectiveness of thrombolytic therapy for acute ischemic stroke. Stroke 2012;43:1315−1322.
    1. Tanne D, Kasner SE, Demchuk AM, et al. Markers of increased risk of intracerebral hemorrhage after intravenous recombinant tissue plasminogen activator therapy for acute ischemic stroke in clinical practice: the Multicenter rt‐PA Stroke Survey. Circulation 2002;105:1679–1685.
    1. Kerr DM, Fulton RL, Lees KR, et al. Seven‐day NIHSS is a sensitive outcome measure for exploratory clinical trials in acute stroke: evidence from the Virtual International Stroke Trials Archive. Stroke 2012;43:1401−1403.
    1. Broderick JP, Lu M, Kothari R, et al. Finding the most powerful measures of the effectiveness of tissue plasminogen activator in the NINDS tPA Stroke Trial. Stroke 2000;31:2335−2341.
    1. Kim JS. tPA helpers in the treatment of acute ischemic stroke: are they ready for clinical use? J Stroke 2019;21:160−174.
    1. Mizuma A, You JS, Yenari MA. Targeting reperfusion injury in the age of mechanical thrombectomy. Stroke 2018;49:1796−1802.
    1. Lo EH. A new penumbra: transitioning from injury into repair after stroke. Nat Med 2008;14:497−500.
    1. Chang JJ, Emanuel BA, Mack WJ, et al. Matrix metalloproteinase‐9: dual role and temporal profile in intracerebral hemorrhage. J Stroke Cerebrovasc Dis 2014;23:2498−2505.
    1. Zhao BQ, Wang S, Kim HY, et al. Role of matrix metalloproteinases in delayed cortical responses after stroke. Nat Med 2006;12:441−445.
    1. Pielecka‐Fortuna J, Kalogeraki E, Fortuna MG, et al. Optimal level activity of matrix metalloproteinases is critical for adult visual plasticity in the healthy and stroke‐affected brain. Elife 2015;26:e11290.
    1. Bencsik P, Pálóczi J, Kocsis GF, et al. Moderate inhibition of myocardial matrix metalloproteinase‐2 by ilomastat is cardioprotective. Pharmacol Res 2014;80:36−42.
    1. Goyal M, Menon BK, van Zwam WH, et al. Endovascular thrombectomy after large‐vessel ischaemic stroke: a meta‐analysis of individual patient data from five randomised trials. Lancet 2016;387:1723−1731.
    1. Nogueira RG, Jadhav AP, Haussen DC, 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.
    1. Albers GW, Marks MP, Kemp S, 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.
    1. Henninger N, Fisher M. Extending the time window for endovascular and pharmacological reperfusion. Trans Stroke Res 2016;7:284−293.

Source: PubMed

3
Tilaa