US20080107641A1 - Method of treating stroke with thrombolytic agent - Google Patents

Method of treating stroke with thrombolytic agent Download PDF

Info

Publication number
US20080107641A1
US20080107641A1 US11/832,291 US83229107A US2008107641A1 US 20080107641 A1 US20080107641 A1 US 20080107641A1 US 83229107 A US83229107 A US 83229107A US 2008107641 A1 US2008107641 A1 US 2008107641A1
Authority
US
United States
Prior art keywords
tenecteplase
bolus
dose
stroke
minutes
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/832,291
Other languages
English (en)
Inventor
Peter Kuebler
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Genentech Inc
Original Assignee
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=38835003&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20080107641(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Genentech Inc filed Critical Genentech Inc
Priority to US11/832,291 priority Critical patent/US20080107641A1/en
Assigned to GENENTECH, INC reassignment GENENTECH, INC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KUEBLER, PETER
Publication of US20080107641A1 publication Critical patent/US20080107641A1/en
Priority to US13/189,413 priority patent/US9023346B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/49Urokinase; Tissue plasminogen activator
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the invention is related to a method of treating stroke with a thrombolytic agent, more particularly, a method of administering tenecteplase in a certain dosing regimen to treat acute ischemic stroke.
  • Stroke is a general term for acute brain damage resulting from disease of the blood vessels. This presents a serious problem to society, with about 500,000 people dying from or becoming permanently disabled by stroke in the United States each year. Stroke can be classified into two main categories: hemorrhagic stroke (resulting from leakage of blood outside of the normal blood vessels) and ischemic stroke (cerebral ischemia due to lack of blood supply); this application is concerned with the latter.
  • Ischemic stroke is responsible for about one third of all deaths in industrialized countries and is the major cause of serious, long-term disability in adults over the age of 45. It stands to reason that there is a need for pharmacotherapy to treat acute ischemic stroke. Considerable insights have been gained into the mechanisms of stroke and the cascade of events that occurs following stroke; there is also an improved understanding of neuronal injury and cell death.
  • ischemic stroke The three main mechanisms of ischemic stroke are thrombosis, embolism, and systemic hypoperfusion (with resultant ischemia and hypoxia).
  • embolism the area of the brain that dies as a result of the lack of blood supply thereto.
  • infarct the area of the brain that dies as a result of the lack of blood supply thereto.
  • Obstruction of a cerebral artery resulting from a thrombus that has built up on the wall of a brain artery is generally called “cerebral thrombosis.”
  • cerebral embolism the occlusive material blocking the cerebral artery arises downstream in the circulation (e.g., an embolus is carried to the cerebral artery from the heart).
  • thromboembolism is used to cover both these types of stroke.
  • Systemic hypoperfusion may arise as a consequence of elevated blood lactate levels, reduced hematocrit, low blood pressure, or inability of the heart to pump blood adequately.
  • TIA transient ischemic attack
  • Acute ischemic stroke is a heterogeneous disease process; prediction of course, recovery, disability, or death is difficult. It is typically due to an acute thromboembolic arterial occlusive lesion. The location of the arterial occlusive lesion in acute ischemic stroke is relatively heterogeneous.
  • Thrombolytic agents such as recombinant tissue plasminogen activator (rtPA) have been used in the treatment of thromboembolic stroke, and function by lysing the thrombus causing the ischemia.
  • rtPA recombinant tissue plasminogen activator
  • intravenous rtPA (alteplase, ACTIVASE®) is the only drug approved for the treatment of acute ischemic stroke.
  • Intravenous rtPA (0.9 mg/kg, maximum 90 mg), with 10% of the dose given as a bolus followed by an infusion lasting 60 minutes, is recommended treatment within 3 hours of onset of ischemic stroke.
  • This drug is believed to be most useful if administered as soon as possible after acute stroke (Gross et al., Neurosurgery, 36:1172-1177 (1995); Ingall et al., Stroke, 35: 2418-2424 (2004); The ATLANTIS, ECASS, and NINDS rt-PA Study Group Investigators, Lancet, 363: 768-774 (2004)), to restore, partially at least, cerebral blood flow in the ischemic region and to sustain neuronal viability.
  • Thrombolysis the lysis of a cerebral arterial clot with tPA within hours of symptom onset in ischemic stroke, has been approved for treatment of acute ischemic stroke since 1996.
  • Two other agents pro-urokinase (intra-arterial administration directly into M1 or M2 arterial thrombus) and intravenous ancrod, a fibrinogen-lowering agent derived from the venom of the Malayan pit viper, have shown therapeutic benefit, and may be available for acute ischemic stroke therapy in the future.
  • pro-urokinase intra-arterial administration directly into M1 or M2 arterial thrombus
  • intravenous ancrod a fibrinogen-lowering agent derived from the venom of the Malayan pit viper
  • 2002/0081294 and US 2004/0057951 disclose co-administration of a thrombolytic compound and an anti-CD 18 antibody for increasing blood flow in an infarct-related artery in a mammal such as a human (e.g., acute myocardial infarction (AMI) in a mammal with a blocked coronary artery or focal ischemic stroke caused by obstruction of a cerebral artery).
  • a mammal such as a human
  • AMI acute myocardial infarction
  • U.S. Pat. No. 6,541,452 discloses a brain-associated inhibitor of tPA and its use in treating stroke.
  • US Pat. Pub. 2004/0176347 discloses a pharmaceutical composition for treating cerebral ischemic diseases comprising an astrocyte-function-improving agent and a thrombolytic agent, preferably tPA, as active ingredients.
  • Tenecteplase (TNK, TNKASETM, Genentech, Inc., South San Francisco, Calif.) is a genetically engineered variant of human tPA cloned and expressed in Chinese hamster ovary cells. Keyt et al., Proc. Natl. Acad. Sci. USA, 91: 3670-3674 (1994). See also Verstraete, Am. J. Med., 109: 52-58 (2000) for an overview of third-generation thrombolytic drugs in general. Approved in the U.S. for a single-bolus administration in patients with AMI, tenecteplase was engineered to have increased fibrin specificity and an increased half-life compared to alteplase.
  • Tenecteplase and alteplase were equivalent for 30-day mortality when single-bolus tenecteplase was compared with front-loaded alteplase in acute myocardial infarction in the ASSENT-2 double-blind randomized trial.
  • the ease of administration of tenecteplase may facilitate more rapid treatment in and out of the hospital. Van de Werf et al., Lancet, 354: 716-722 (1999).
  • the results of the ASSENT-2 study indicated that total stroke rate and 30-day mortality were lower in female patients over 75 years of age treated with tenecteplase than in those treated with alteplase, albeit that the difference was statistically not significant.
  • thrombolytic agents that is given according to a weight-adjusted dose regimen, e.g., tenecteplase. Vermeer, Thrombosis Research, 103: Supplement 1, S101-S104 (Sep. 30, 2001).
  • Other thrombolytic drugs that may be useful in treating AMI include streptokinase, urokinase, anistreplase,reteplase, saruplase, reteplase, lanoteplase, staphylokinase, fibrolase, prourokinase, and vampire bat plasminogen activator.
  • Tenecteplase was found to be effective in treating AMI in combination with the low-molecular-weight heparin enoxaparin (ENOX) or unfractionated heparin in the prehospital setting in a trial called ASSENT-3 PLUS.
  • ENOX low-molecular-weight heparin enoxaparin
  • ASSENT-3 PLUS unfractionated heparin in the prehospital setting in a trial called ASSENT-3 PLUS.
  • the combination of tenecteplase with ENOX reduces early ischemic events, but lower doses of ENOX need to be tested in elderly patients.
  • the animal data also show that, overall, the beneficial effects of the neuroprotective agents are weaker, and are totally lost sooner, than those of thrombolytics.
  • the human data show that the failed trials of the neuroprotective agents had entry windows that went far beyond the windows of (any) success seen in tests of these agents in animals.
  • human thrombolysis trials uniformly restricted time of entry to windows in which these agents have shown beneficial effect in animals.
  • neuroprotective agents failed to produce benefit because their effects at best are too weak, and they were used at times predictable from the animal models as too late.
  • Thrombolytic therapy such as tenecteplase and urokinase, which has a stronger effect than neuroprotective agents in animal models, was used clinically during the early window of optimal effectiveness, and produced beneficial results.
  • third-generation thrombolytics Two of the several third-generation thrombolytic agents have been investigated for the treatment of acute ischemic stroke and include tenecteplase and reteplase. By virtue of structural modifications, third-generation thrombolytics have longer half-lives and greater penetration into the thrombus matrix.
  • the first prospective human clinical trial evaluated the safety and efficacy of intra-arterial reteplase in 16 patients with ischemic stroke who were poor candidates for intravenous alteplase therapy.
  • monteplase a modified rtPA
  • Muramatsu et al. Neurological Research, 24: 311-316 (2002).
  • Other such third-generation drugs include lanoteplase, plasmin, or a truncated form of plasmin (microplasmin), a direct-acting thrombolytic with non-thrombolytic-related neuroprotective, therapeutic activities, recombinant desmodus rotundus salivary plasminogen activator (rDSPA) alpha-1, and a mutant fibrin-activated human plasminogen (BB10153; British Biotech Inc.).
  • CLEAR stroke study A multi-center, randomized, double-blinded sequential dose-escalation clinical trial called the CLEAR stroke study is now being conducted to evaluate the safety of eptifibatide, an intravenous cyclical heptapeptide that selectively blocks the platelet glycoprotein IIb/IIIa receptor, in combination with low-dose rtPA in acute ischemic stroke treated within three hours.
  • tenecteplase may be neuroprotective following a stroke because of its increased fibrin specificity over alteplase, its resistance to PAI-1, and its increased biological half-life (18 vs. 10 minutes for alteplase), features that could lead to fewer cerebral hemorrhages than alteplase in stroke patients.
  • tenecteplase A pilot study of tenecteplase was made in 88 acute ischemic stroke patients enrolled over 2000 to 2003 using four dose tiers of tenecteplase: 0.1, 0.2, 0.4, and 0.5 mg/kg. There were no symptomatic intracranial hemorrhages (ICHs) in the first three tiers. Two of 13 patients had symptomatic ICH at 0.5 mg/kg, and there were increasing ICHs with increasing doses (8%-38%), with outcomes similar to the alteplase group in the earlier acute ischemic stroke trial.
  • ICHs intracranial hemorrhages
  • Tenecteplase is currently being tested in a randomized controlled Phase IIb clinical study in acute ischemic stroke patients using 0.1 mg/kg tenecteplase, 0.4 mg/kg tenecteplase, and 0.9 mg/kg rtPA.
  • tenecteplase In an early animal study, the activity of tenecteplase was compared with that of alteplase in rabbit models of embolic stroke and peripheral bleeding. Infusion of alteplase or bolus administration of the tenecteplase resulted in dose-dependent clot lysis. The tenecteplase was found to be an order of magnitude more potent than alteplase on a milligram-per-kilogram basis. Unlike alteplase, tenecteplase caused less systemic activation of plasminogen and fewer hemorrhagic transformations in this model. The tenecteplase did not extend template bleeding times.
  • tenecteplase in a dose of using 0.6 mg/kg or 1.5 mg/kg was compared with wild-type tPA in a rabbit embolic stroke model. Both wild-type tPA and tenecteplase caused thrombolysis in most subjects, and did not differ from each other. Neither tenecteplase nor tPA affected the size of the hemorrhages. Tenecteplase shows comparable rates of recanalization compared with wild-type tPA in a model of embolic stroke. While tPA increases hemorrhage rate, the hemorrhage associated with tenecteplase treatment is not statistically different compared with controls or the tPA group.
  • 188(2) 279-85(2004) discloses the window for acute stroke treatment of thrombolytics such as tenecteplase plus central-nervous-system (CNS)-protective therapies such as free-radical scavengers, NXY 059, and nitrogen oxides.
  • CNS central-nervous-system
  • Lapchak et al., Experimental Neurology, 185: 154-159 (2004) discloses a comparison of tenecteplase with alteplase on clinical rating scores following small-clot embolic strokes in rabbits.
  • the rabbit small clot embolic stroke model (RSCEM) was used for a dose-response profile analysis of tenecteplase (0.1 mg/kg-3.3 mg/kg) and alteplase (0.9 mg/kg-3.3 mg/kg) given intravenously 1 hour following embolization.
  • tenecteplase (0.9 mg/kg) or alteplase (3.3 mg/kg) was administered 3 (or 6) hours following embolization to determine the therapeutic window for the thrombolytics.
  • behavioral analysis was conducted 24 hours following embolization, allowing for the determination of the effective stroke dose (P50) or clot amount (mg) that produces neurological deficits in 50% of the rabbits.
  • tenecteplase has a wide therapeutic range, a therapeutic window of at least 3 hours, and a durable effect. Moreover, the safety profile for tenecteplase is similar to that of alteplase. Tenecteplase does not increase the rate of intracerebral hemorrhage (ICH) above that produced by alteplase. However, the therapeutic range and window for alteplase is more limited than that for tenecteplase. These preclinical studies suggest that tenecteplase has a better pharmacological profile than alteplase and supports further investigation of tenecteplase in randomized double-blinded clinical trials in stroke patients.
  • the invention provides a method for treating acute ischemic stroke in a human comprising administering tenecteplase to the human in a total dose of about 0.05 to 0.5 mg/kg, given as (a) an initial bolus dose of about 0.015 to 0.15 mg/kg, followed by infusion of an amount equaling the total dose minus the initial dose over a period of about 50-90 minutes, or (b) a bolus only.
  • tenecteplase is administered to the human in the form of a pharmaceutically acceptable formulation, such as those elaborated in more detail herein.
  • the total dose is about 0.2 to 0.3 mg/kg, more preferably about 0.25 mg/kg.
  • the total dose is given as an initial bolus followed by the infusion.
  • the initial dose is about 0.08 to 0.12 mg/kg, more preferably about 0.1 mg/kg bolus, and/or the period of infusion is about 55-70 minutes, more preferably about 60 minutes.
  • the total dose is about 0.25 mg/kg, given as an initial about 0.1 mg/kg bolus, followed by infusion of about 0.15 mg/kg over about 60 minutes.
  • tenecteplase is administered to the human in a total dose of about 0.25 mg/kg in about 60 minutes, given as an initial bolus of about 0.1 mg/kg over one minute, followed by infusion of about 0.25 mg/kg for the rest of about 60 minutes.
  • the total dose is given as a bolus only.
  • the total dose typically is about 0.05 to about 0.5 mg/kg. In a preferred embodiment, the total dose is about 0.25 mg/kg.
  • the tenecteplase is administered to the human at a time between about 15 minutes to about 20 hours from the onset of acute ischemic stroke, more preferably between about 45 minutes to about 6 hours, and still more preferably up to no more than about 3 hours from the onset of acute ischemic stroke.
  • the bolus is intravenous and/or the infusion is continuous.
  • both these methods further comprise administering to the human an effective amount of a second medicament, wherein the first medicament is tenecteplase.
  • This second medicament is preferably a neuroprotective agent, a thrombolytic agent, a glycoprotein IIb IIIa antagonist, or an anti-CD18 antibody.
  • This second medicament may be co-administered to the human either before, after, or simultaneously with, the tenecteplase.
  • Such second medicament may be administered to the mammal more than about 3 hours after the onset of ischemic stroke (e.g., at least once within about 3-8 hours and preferably within about 3-6 hours from the onset of stroke).
  • the invention supplies a kit comprising: a container comprising tenecteplase; and instructions for using the tenecteplase to treat acute ischemic stroke in a human by administering the tenecteplase to the human in a total dose of about 0.05 to 0.5 mg/kg, given as (a) an initial bolus dose of about 0.015 to 0.15 mg/kg, followed by infusion of an amount equaling the total dose minus the initial dose over a period of about 50-90 minutes, or (b) a bolus.
  • the total dose is about 0.2 to 0.3 mg/kg, more preferably about 0.25 mg/kg, and the initial bolus is about 0.08 to 0.12 mg/kg.
  • the total dose is given as an initial bolus followed by the infusion.
  • the total dose is about 0.25 mg/kg, given as an initial about 0.1 mg/kg bolus, followed by infusion of about 0.15 mg/kg over 60 minutes.
  • the total dose is about 0.25 mg/kg, given as an initial about 0.1 mg/kg bolus, followed by infusion of about 0.15 mg/kg over about 60 minutes.
  • tenecteplase is given in a total dose of about 0.25 mg/kg in about 60 minutes, given as an initial bolus of about 0.1 mg/kg over one minute, followed by infusion of about 0.25 mg/kg for the rest of 60 minutes.
  • the total dose is given as a bolus only.
  • kits herein preferably further comprise a container comprising a second medicament, wherein the instructions include instructions for using the second medicament in combination with the tenecteplase to treat ischemic stroke in a human by administering to the human an effective amount of the second medicament.
  • the preferred second medicament is a neuroprotective agent, a thrombolytic agent, a glycoprotein IIb IIIa antagonist, or an anti-CD18 antibody.
  • FIG. 1 shows median model-predicted ACTIVASE® (alteplase) concentrations after 0.9 m/kg as a 10% bolus over 1 minute and 90% over the remainder of 1 hour. These were the results of a 1000-subject simulation. Demographics were based on those observed in a pilot AIS stroke study.
  • FIG. 2 shows model-predicted pharmacokinetic (PK) profiles from 1000-subject simulations comparing the median model-predictedreteplase (ACTIVASE®) concentrations with the model-predicted tenecteplase (TNK) median, 5 th , and 95 th percentile concentrations from a 0.25 mg/kg bolus-infusion regimen.
  • the simulation was based on TNKase administered as 0.25 mg/kg as 0.1 mg/kg bolus over 1 minute, 0.15 mg/kg over remainder of one hour.
  • TNK 5 th Percentile is the lowest (and solid) line on the graph
  • TNK Median is the dotted line close to the Activase Median line
  • TNK 95 th Percentile is the uppermost dashed line.
  • the median model-predicted alteplase concentration was based on 0.9 mg/kg (90 mg/kg max) as 10% bolus and 90% over the remainder of one hour (Activase Median is the solid line that plateaus and then drops off after 60 minutes).
  • FIG. 3 shows model-predicted PK profiles from 1000-subject simulations comparing the median model-predictedreteplase (ACTIVASE®) concentrations for a bolus-infusion regimen with the model-predicted tenecteplase (TNK) median, 5 th , and 95 th percentile concentrations from a 0.25 mg/kg bolus-only regimen.
  • TNK 5 th Percentile is the lowest (and solid) line on the graph
  • TNK Median is the dotted line above the Activase Median line
  • TNK 95 th Percentile is the uppermost dashed line.
  • the median model-predicted alteplase concentration was based on 0.9 mg/kg (90 mg/kg max) as 10% bolus and 90% over the remainder of one hour (Activase Median is the solid line that plateaus and then drops off after 60 minutes).
  • Stroke is defined herein as a neurologic deficit caused by a cerebrovascular accident (CVA), which disrupts the blood supply to the brain for at least 24 hours. Stroke may take different forms, including hemorrhagic stroke and ischemic stroke, where each may be further subdivided. Thus, for example, hemorrhagic stroke may be characterized by a sudden development of neurological deficit with ICH or subarachnoid hemorrhage (SAH), while subtypes of ischemic stroke include lacunar, thromboembolic, and cardioembolic strokes.
  • SAH subarachnoid hemorrhage
  • ischemic stroke include lacunar, thromboembolic, and cardioembolic strokes.
  • stroke is used herein in the broadest sense, and includes all forms of stroke, whether specifically listed herein or not.
  • Transient ischemic attack or “TIA” is defined herein as a temporary disruption in the blood supply to the brain, which is resolved completely within 24 hours, and usually lasts minutes to an hour.
  • Acute ischemic stroke is defined herein as an acute development of focal or global disturbance of cerebral function due to thromboembolism lasting more than 24 hours or leading to death.
  • An acute focal ischemic stroke is damage to the brain caused by interruption of the blood supply to a region thereof.
  • the acute ischemic stroke of interest herein is generally caused by obstruction of any one or more of the arteries, including the main cerebral arteries (e.g., middle cerebral artery, anterior cerebral artery, posterior cerebral artery, internal carotid artery, vertebral artery or basilar artery), and secondary arteries or arterioles.
  • the “arterial obstruction” is generally a single embolus or thrombus or a plurality of clot particles that occlude primary and secondary arteries or arterioles.
  • IVH intraventricular hemorrhage
  • grade 1 bleeding occurs in a small area of the ventricles
  • grade 2 bleeding also occurs inside of the ventricles
  • grade 3 ventricles are enlarged by the blood
  • grade 4 bleeding extends into the brain tissue around the ventricles.
  • increasing cerebral blood flow or reducing infarct size is meant the act of improving clinical outcome by inducing a statistically or physiologically significant increase in cerebral blood flow and/or a statistically or physiologically significant reduction in infarct size in a treated mammal relative to an untreated mammal as determined using techniques that are well known in the art, such as vascular imaging, for example.
  • cerebral blood flow as determined 2-4 hours after administration of the antagonist is increased by at least 30% and preferably at least 50% relative to an untreated mammal.
  • infarct size measured 48 hours after administration of the antagonist will be 20% less and preferably 50% less than that of an untreated mammal.
  • infarct is an area of necrosis in a tissue or organ, for example, heart or brain, resulting from obstruction of the local circulation by a thrombus or embolus. Infarct size can be measured by known methods.
  • an “infarct-related artery” is an artery that, when at least partially blocked by a thrombus or embolus, gives rise to an infarct in a tissue or organ, for example, heart or brain.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder as well as those in which the disorder is to be prevented. Preferred herein is the treatment of those individuals who have been diagnosed as having suffered stroke, in particular, acute ischemic stroke.
  • tenecteplase also known as TNK-tPA or TNKASETM brand of tissue-plasminogen activator variant, refers to a tPA variant designated T103N, N117Q, K296A, H297A, R298A, R299A tPA available from Genentech, Inc.
  • a “package insert” is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications, other therapeutic products to be combined with the packaged product, and/or warnings concerning the use of such therapeutic products, etc.
  • a “medicament” is an active drug to treat stroke or its symptoms or side effects.
  • a “second medicament” is one that can be added to help the first medicament, tenecteplase, to treat the stroke.
  • second medicaments include, without limitation, aspirin, intercellular adhesion molecule (ICAM)-1 and LFA-1 antagonists including antibodies such as enlimomab (an anti-ICAM-1 monoclonal antibody), and anti-CD18 and anti-CD11a antibodies, human anti-leukocytic antibodies such as Hu23F2G, glycoprotein IIb IIIa antagonists such as eptifibatide (INTEGRELINTM) (an intravenous cyclical heptapeptide that selectively blocks the platelet glycoprotein IIb/IIIa receptor), direct thrombin inhibitors, external or local ultrasound, mechanical clot retrieval or inaceration, fibrinolytic agents, neuronal wound healing agents such as basic fibroblast growth factor (e.g., FIBLASTTM), neuroprotective agents such as citicoline, magnesium, nalmefene, dizocilpine
  • a “thrombolytic agent” is a molecule that breaks up and/or dissolves a thrombus.
  • exemplary thrombolytic agents include streptokinase, acylated plasminogen-streptokinase activator complex (APSAC), urokinase, single-chain urokinase-plasminogen activator (scu-PA), thrombin-like enzymes from snake venoms such as ancrod (Bell, W. “Defibrinogenating enzymes” In Colman et al., (eds): Hemostasis and Thrombosis Lippincott, Philadelphia (1987) p. 886), tPA, and biologically active variants of each of the above.
  • Suitable thrombolytic agents that may be used in this invention are disclosed, for example, in U.S. Pat. Nos. 5,770,425; 5,770,426; 5,612,029; 5,520,911; 5,736,134; 5,728,567; 5,714,145; 5,840,564; 5,616,486; 5,411,871; 5,520,913; 5,262,170; and 5,108,901.
  • Co-administration means the administration of the second medicament during the effective therapeutic window of the tenecteplase administered alone.
  • the second medicament may be administered before, concurrent with, or after the tenecteplase.
  • the administration of the second medicament such as anti-CD18 antibody, is preferably started from about 1 hour before up to immediately (1-15 minutes) before, more preferably concurrently with, the start of administration of the tenecteplase.
  • Co-administration also encompasses administration of the second medicament after the start of administration of the tenecteplase, for example about 15-30 minutes after and up to about 3 hour after.
  • Co-administration includes administration in the form of a single formulation, where the two medicaments may be, but do not have to be, physically separated from each other.
  • the “effective therapeutic window” of tenecteplase administered alone means the time period or time window following an infarct caused by blockage of an artery during which the tenecteplase, when administered alone, is effective in reestablishing patency of blood flow in the artery relative to a control not receiving the tenecteplase.
  • the effective therapeutic window is species dependent for tenecteplase, but can be readily determined by standard tests evaluating the efficacy of the tenecteplase versus controls.
  • anti-CD18 antibody when used herein refers to an antibody that binds to CD18 (preferably human CD18) and inhibits or substantially reduces a biological activity of CD18. Normally, the antibody will block (partially or completely) the ability of a cell (e.g., a neutrophil) expressing the CD18 subunit at its cell surface to bind to endothelium.
  • a cell e.g., a neutrophil
  • anti-CD18 antibodies examples include MHM23 (Hildreth et al. (1983)); M18/2 (IgG 2a ; Sanches-Madrid et al., Eur. J. Immunol. 13(3):202-208 (1983)); H52 (American Type Culture Collection (ATCC) Deposit HB10160); Mas191c and IOT18 (Vermot Desroches et al., Scand. J. Immunol. 33(3):277-286 (1991)); and NA-8 (WO 94/12214).
  • the preferred antibody is one that binds to the CD18 epitope to which either MHM23 or H52 binds.
  • the antibody has a high affinity for the CD18 polypeptide.
  • the antibody has an affinity for the CD18 antigen of about 4 nM or less.
  • the affinity is about 3 nM or less, and most preferably about 1 nM or less.
  • the antibody may bind to a region in the extracellular domain of CD18 that associates with CD11b and the antibody may also dissociate alpha and beta chains (e.g., the antibody may dissociate the CD11b and CD18 complex, as is the case for the MHM23 antibody).
  • the present invention provides an improved protocol for the treatment of stroke, in particular acute ischemic stroke, with tenecteplase.
  • the treatment protocols and dosing regimens of the present invention result in pharmacokinetic profiles that offer maximum efficacy and safety, and thus represent a significant advance in the thrombolytic therapy of stroke.
  • the invention provides a method for treating acute ischemic stroke in a human comprising administering to the human tenecteplase in a total dose of about 0.05 to 0.15 mg/kg (preferably about 0.2 to 0.3 mg/kg, and more preferably about 0.25 mg/kg), given as an initial dose of about 0.015 to 0.15 mg/kg bolus (preferably about 0.08 to 0.12 mg/kg bolus, more preferably about 0.1 mg/kg bolus), followed by infusion of the remaining amount to total about 0.05 to 0.5 mg/kg (preferably about 0.2 to 0.3 mg/kg, more preferably about 0.25 mg/kg) over a period of about 50-90 minutes, more preferably about 55-70 minutes, and most preferably about 60 minutes.
  • the initial bolus dose is preferably about 0.1 mg/kg and the subsequent infusion is about 0.15 mg/kg. Based on current experiments, this is the most preferred regimen, wherein the subsequent infusion is given over about 60 minutes. It is noted, however, that the most preferred dosing schedule might vary within the specified dosing ranges, depending on various factors, including the specific type and extent of stroke, the condition of the patient, the time elapsed from the onset of stroke, and the like.
  • the infusion of tenecteplase may follow immediately after the bolus dose is complete, or can be separated from completion of the bolus dosage by up to about 30 minutes, but it is preferred to initiate the infusion immediately after the bolus dose is completed.
  • the bolus injection is intravenous, but it may be injected by other means such as intra-arterially.
  • the infusion is continuous by intravenous, intramuscular, intraperitoneal, intracerebrospinal, subcutaneous, intra-articular, intrasynovial, or intrathecal routes, but the preferred infusion is intravenous.
  • acute ischemic stroke is treated in a human by administering tenecteplase in a total dose of about 0.05 to 0.15 mg/kg, preferably about 0.2 to 0.3 mg/kg, more preferably about 0.25 mg/kg, given exclusively as a bolus dose.
  • the bolus is intravenous.
  • Stroke is a serious condition and the third leading cause of death in the United States. Since survival and the extent of recovery are a function of the time of diagnosis and intervention, in the methods of the present invention it is contemplated that the tenecteplase will be administered to a patient as soon as possible once the condition of acute ischemic stroke has been diagnosed or is suggested by acute deficit on neurologic examination.
  • Initial clinical presentations of acute ischemic stroke typically include one or more of (1) alterations in consciousness, such as stupor or coma, confusion or agitation, memory loss, seizures, and/or delirium; (2) headache that is intense or unusually severe, is associated with decreased level of consciousness/neurological deficit, and/or includes unusual/severe neck or facial pain; (3) aphasia (incoherent speech or difficulty understanding speech); (4) facial weakness or asymmetry; (5) uncoordination, weakness, paralysis, or sensory loss of one or more limbs; (6) ataxia (poor balance, clumsiness, or difficulty walking); (7) visual loss; and (8) intense vertigo, double vision, unilateral hearing loss, nausea, vomiting and/or photophobia.
  • the presence of one or more of these manifestations might be an initial indication of acute ischemic stroke, which can be verified by follow-up differential diagnosis and neurological examination.
  • Neurologic examination and, optionally, neuroimaging techniques such as computed tomography (CT) (including non-contrast CT and perfusion CT) and magnetic resonance imaging (MRI) (including diffusion weighted imaging (DWI) and perfusion imaging (PI)); vascular imaging (e.g., duplex scanning and transcranial Doppler ultrasound and laser Doppler); and angiography (e.g., computerized digital subtraction angiography (DSA) and MR angiography) as well as other invasive or non-invasive techniques, are available for the diagnosis of acute ischemic stroke.
  • CT computed tomography
  • MRI magnetic resonance imaging
  • DWI diffusion weighted imaging
  • PI perfusion imaging
  • vascular imaging e.g., duplex scanning and transcranial Doppler ultrasound and laser Doppler
  • angiography e.g., computerized digital subtraction angiography (DSA) and MR angiography
  • DSA computerized digital subtraction angiography
  • NIHSS National Institute of Health Stroke Scale
  • the tenecteplase will be administered in the dosage and dosage regimen herein at least once at any time from immediately following to about 24 hours after the onset of stroke.
  • the tenecteplase is first administered to the patient between about 15 minutes (or about 30 or 45 minutes) to about 20 hours (more preferably about 10 hours, or about 6 hours, or 3 hours, or about 90 minutes, or about 60 minutes) from the onset of stroke.
  • a patient presenting within 3 hours of the onset of signs and symptoms consistent with an acute ischemic stroke is subjected to thrombolytic therapy with tenecteplase in accordance with the present invention.
  • the tenecteplase is a first medicament
  • the second medicament may be one or more medicaments, and may include, for example, those set forth above.
  • Preferred such medicaments include neuroprotective agents, anticonvulsive agents, a spin-trap agent, intercellular adhesion molecule (ICAM)-1 and LFA-1 antagonists such as anti-CD11a and anti-CD18 antibodies, glycoprotein IIb IIIa antagonists, neuronal wound healing agent, antibodies inhibiting platelet aggregation and adhesion, and human anti-leukocytic antibodies, or another thrombolytic agent than tenecteplase. More preferred are neuroprotective agents, other thrombolytic agents, glycoprotein IIb IIIa antagonists, and anti-CD18 antibodies.
  • second medicaments are generally used in the same dosages and with administration routes as used hereinbefore or about from 1 to 99% of the heretofore-employed dosages. If such second medicaments are used at all, preferably, they are used in lower amounts than if the tenecteplase were not present, especially in subsequent dosings beyond the initial dosing with tenecteplase, so as to eliminate or reduce side effects caused thereby.
  • a second medicament is administered in an effective amount with a tenecteplase bolus dosing, it may be administered with any such dosing, for example, only with one such dosing, or with more than one such dosing.
  • the second medicament is administered with the initial bolus dosing.
  • the second medicament is administered with the first and second dosings.
  • the second medicament is administered with all tenecteplase dosings.
  • the combined administration includes co-administration (concurrent administration), using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents simultaneously exert their biological activities. It is preferred that after the initial exposure, the amount of such agent is reduced or eliminated so as to reduce the exposure of the subject to an agent with side effects such as prednisone and cyclophosphamide.
  • a device such as an INTERCOOLTM device and/or using external ice at 33° C. or a similar temperature may be employed along with the tenecteplase for treating the stroke.
  • Therapeutic formulations of the tenecteplase are prepared for storage by mixing the tenecteplase having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate and other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin or immunoglobulins; amino acids such as glycine, glutamine, asparagine, histidine, arginine or lysine; monosaccharides, disaccharides and other carbohydrates including glucose, mannose, trehalose or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEENTM, PLURONICSTM or PEG.
  • buffers such as phosphate, citrate and other organic acids
  • antioxidants including ascorbic acid
  • low molecular weight (less than about 10 residues) polypeptides proteins
  • the active ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • the formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes, prior to or following lyophilization and reconstitution.
  • sustained-release preparations may be employed. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the tenecteplase, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and y ethyl-L-glutamate non-degradable ethylene-vinyl acetate
  • degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate)
  • poly-D-( ⁇ )-3-hydroxybutyric acid While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods.
  • encapsulated antibodies When encapsulated antibodies remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37° C., resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S—S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
  • Sustained-release tenecteplase compositions also include liposomally entrapped tenecteplase.
  • Liposomes containing the tenecteplase are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA, 82:3688 (1985); Hwang et al., Proc. Natl. Acad. Sci. USA, 77:4030 (1980); and U.S. Pat. Nos. 4,485,045 and 4,544,545.
  • the liposomes are the small (about 200-800 Angstroms) unilamelar type in which the lipid content is greater than about 30 mol. % cholesterol, the selected proportion being adjusted for the optimal tenecteplase therapy. Liposomes with enhanced circulation time are disclosed in U.S. Pat. No. 5,013,556.
  • tenecteplase The exact total dosage of tenecteplase to be employed, and how much is by bolus and how much by infusion, or whether only bolus should be employed, will depend, for example, on the exact nature of the stroke to be treated, the severity and course of the stroke, whether the tenecteplase is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the tenecteplase, and the discretion of the attending physician. The progress of this therapy is easily monitored by conventional techniques and assays elaborated herein.
  • the article of manufacture comprises a container with a label.
  • Suitable containers include, for example, bottles, vials, and test tubes.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition that is effective for treating stroke as defined herein and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the active agent in the composition is tenecteplase.
  • the label on the container indicates that the composition is used for treating stroke as described above, and may also indicate directions for in vivo use, such as those described above.
  • the kit comprises a container comprising tenecteplase and instructions for using the tenecteplase to treat acute ischemic stroke in a human by administering the tenecteplase to the human in a total dose of about 0.05 to 0.5 mg/kg, given as (a) an initial bolus dose of about 0.015 to 0.15 mg/kg, followed by infusion of an amount equaling the total dose minus the initial dose over a period of about 50-90 minutes, or (b) a bolus.
  • the total dose is about 0.2 to 0.3 mg/kg, more preferably about 0.25 mg/kg
  • the initial dose under option (a) above is about 0.08 to 0.12 mg/kg, more preferably about 0.1 mg/kg.
  • the total dose is given as an initial bolus followed by the infusion.
  • the total dose is about 0.25 mg/kg, given as an initial about 0.1 mg/kg bolus, followed by infusion of about 0.15 mg/kg over about 60 minutes.
  • the total dose is given as a bolus.
  • kits may optionally also comprise a container holding a second medicament, wherein the instructions include directions for using the second medicament in combination with the tenecteplase to treat ischemic stroke in a human by administering to the human an effective amount of the second medicament.
  • the instructions include directions for using the second medicament in combination with the tenecteplase to treat ischemic stroke in a human by administering to the human an effective amount of the second medicament.
  • Exemplary second medicaments and preferred second medicaments are noted above.
  • kits of the invention may also comprise another container comprising a pharmaceutically acceptable buffer, such as phosphate-buffered saline, Ringer's solution, and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • a pharmaceutically acceptable buffer such as phosphate-buffered saline, Ringer's solution, and dextrose solution.
  • a dosing strategy that improves the safety and efficacy of the treatment of AIS with tenecteplase was developed by performing PK modeling.
  • PK analysis of a rabbit in-vivo PK and pharmacodynamic (PD) study suggested that alteplase and TNKase were approximately equipotent when comparing the area under the curve (AUC) and time to 50% clot lysis (Thomas et al. Stroke, 25/10 2072-2078 (1994)).
  • AUC area under the curve
  • Thomas et al. Stroke, 25/10 2072-2078 (1994) was also inferred from the TIMI10B clinical study of AMI patients treated with alteplase and TNKase, where not only the efficacy and safety outcomes were found to be similar, but also the concentrations of alteplase and TNKase at the key treatment time-points of 30 minutes and 90 minutes.
  • PK modeling was performed using historical alteplase acute myocardial infarct (AMI) PK data as well as TNKase data in AMI and stroke to an provide appropriate dose of TNKase for AIS.
  • AMI acute myocardial infarct
  • a bolus of 0.25 mg/kg bolus was the 2nd choice for a TNKase dose in AIS dosing strategy.
  • the TNKase dosing regimens were derived using a modeling and simulation approach intended to recapitulate the exposure and concentration time profile of alteplase during the treatment period associated with the USPI dosing of 0.9 mg/kg (10% bolus over 1 minute and 90% over the remainder of an hour).
  • PK model an alteplase structural and error model
  • PPK model TNKase population PK model
  • TNKase PPK model was derived using individual patient serum concentration data from the AMI trial TIMI10B (Modi et al., supra) and a pilot dose-escalating AIS trial. The combined data were best described using a two-compartment model. The final model results are summarized in Table II. TABLE II TNKase (Tenecteplase) Final Model Estimates Model Result Parameter Parameter estimate (SEE) Method FOCE with INTER No.
  • the basis for the TNKase dosing regimens was the concentration-time profile and exposure of the USPI dosing regimen for alteplase during the 60-minute treatment period ( FIG. 1 ).
  • the strategy was to use this as an approximate target for TNKase dosing.
  • FIG. 1 suggested that the 10% alteplase bolus results in near steady-state drug concentrations, followed by a fairly constant concentration of alteplase for the remaining 60 minutes.
  • Model-estimated median concentration values obtained at two-minute intervals from 2-60 minutes were then used to calculate mean effective concentration (computer program JMP version 5.1.2003) (SAS Institute Inc.) and the AUC.
  • the first TNKase regimen for stroke included a bolus followed by a constant infusion.
  • the mean effective concentration for alteplase was approximately 1800 ng/mL, resulting in an AUC of 108,000 ng*min/mL.
  • the total TNKase dose required to maintain a concentration and exposure approximating the effective concentration was 0.16 mg/kg administered over 1 hour.
  • FIG. 2 shows how the mean, 5 th , and 95 th percentile concentrations from this regimen compared to the model-predicted concentrations for alteplase.
  • TNKase a dose of TNKase of 0.25 mg/kg administered as 0.1 mg/kg bolus over 1 minute and 0.15 mg/kg over the remainder of 1 hour was determined as an appropriate dosing regimen for TNKase in AIS.
  • the intent was to administer higher early doses to improve clot lysis (consistent with what has been observed with alteplase in AMI) without altering fibrinogen or increasing the risk of bleeding.
  • This bolus regimen also would reduce the low-level exposure at later time points observed with the bolus-infusion regimen.
  • FIG. 3 shows the median model-predicted alteplase concentrations with the model-predicted TNKase median, 5 th , and 95 th percentile concentrations from the 0.25 mg/kg bolus regimen.
  • Table IV summarizes the AUC values and compares concentrations at key timepoints. Overall, the exposure and concentration time curves observed from simulating a 0.25 mg/kg bolus suggested that this regimen for bolus-only treatment was appropriate.
  • bolus-infusion and bolus-only dosing regimens of TNKase for use in treating acute ischemic stroke were determined using pharmacokinetic modeling and simulation. These dosing regimens were designed to provide improved safety and efficacy from tenecteplase as compared to alteplase when administered to AIS patients.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Dermatology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
US11/832,291 2006-08-29 2007-08-01 Method of treating stroke with thrombolytic agent Abandoned US20080107641A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/832,291 US20080107641A1 (en) 2006-08-29 2007-08-01 Method of treating stroke with thrombolytic agent
US13/189,413 US9023346B2 (en) 2006-08-29 2011-07-22 Method of treating stroke with tenecteplase

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US82386806P 2006-08-29 2006-08-29
US11/832,291 US20080107641A1 (en) 2006-08-29 2007-08-01 Method of treating stroke with thrombolytic agent

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/189,413 Continuation US9023346B2 (en) 2006-08-29 2011-07-22 Method of treating stroke with tenecteplase

Publications (1)

Publication Number Publication Date
US20080107641A1 true US20080107641A1 (en) 2008-05-08

Family

ID=38835003

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/832,291 Abandoned US20080107641A1 (en) 2006-08-29 2007-08-01 Method of treating stroke with thrombolytic agent
US13/189,413 Active 2029-01-12 US9023346B2 (en) 2006-08-29 2011-07-22 Method of treating stroke with tenecteplase

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/189,413 Active 2029-01-12 US9023346B2 (en) 2006-08-29 2011-07-22 Method of treating stroke with tenecteplase

Country Status (22)

Country Link
US (2) US20080107641A1 (ja)
EP (1) EP2056856B1 (ja)
JP (1) JP5785687B2 (ja)
KR (1) KR20090045405A (ja)
CN (1) CN101505785A (ja)
AU (1) AU2007290277B2 (ja)
BR (1) BRPI0716143A2 (ja)
CA (1) CA2661012C (ja)
CO (1) CO6150188A2 (ja)
EA (1) EA015573B1 (ja)
EC (1) ECSP099178A (ja)
HK (1) HK1131054A1 (ja)
IL (2) IL196987A (ja)
MX (1) MX2009001918A (ja)
MY (1) MY163119A (ja)
NO (1) NO342302B1 (ja)
NZ (1) NZ574767A (ja)
SG (3) SG174753A1 (ja)
TW (1) TWI439282B (ja)
UA (1) UA97486C2 (ja)
WO (1) WO2008027687A2 (ja)
ZA (1) ZA200900957B (ja)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011028503A1 (en) * 2009-08-24 2011-03-10 Hough Ear Institute Methods for treating acute acoustic trauma
US20110257510A1 (en) * 2008-12-24 2011-10-20 Koninklijke Philips Electronics N.V. System, method and apparatus for cardiac intervention with mr stroke detection and treatment
US8679150B1 (en) 2013-03-15 2014-03-25 Insera Therapeutics, Inc. Shape-set textile structure based mechanical thrombectomy methods
US8690907B1 (en) 2013-03-15 2014-04-08 Insera Therapeutics, Inc. Vascular treatment methods
US8715317B1 (en) 2013-07-29 2014-05-06 Insera Therapeutics, Inc. Flow diverting devices
US9023346B2 (en) 2006-08-29 2015-05-05 Genentech, Inc. Method of treating stroke with tenecteplase
US9034007B2 (en) 2007-09-21 2015-05-19 Insera Therapeutics, Inc. Distal embolic protection devices with a variable thickness microguidewire and methods for their use
US20150238502A1 (en) * 2012-10-18 2015-08-27 University Of South Florida Compositions and Methods for Treating Stroke
US9314324B2 (en) 2013-03-15 2016-04-19 Insera Therapeutics, Inc. Vascular treatment devices and methods
US9943575B2 (en) * 2010-12-23 2018-04-17 Gennova Biopharmaceuticals Limited Pharmaceutical compositions of tenecteplase
US10390926B2 (en) 2013-07-29 2019-08-27 Insera Therapeutics, Inc. Aspiration devices and methods
US10426424B2 (en) 2017-11-21 2019-10-01 General Electric Company System and method for generating and performing imaging protocol simulations
WO2021158799A1 (en) * 2020-02-04 2021-08-12 The Regents Of The University Of Colorado, A Body Corporate Prophylactic uses of annexin a2

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7759506B2 (en) 2002-02-25 2010-07-20 Diffusion Pharmaceuticals Llc Bipolar trans carotenoid salts and their uses
UA95903C2 (ru) 2005-02-24 2011-09-26 Дифьюжен Фармасьютикалз Ллк Транс-каротиноиды, их синтез, лекарственная форма и применение
JP2010524855A (ja) 2007-04-13 2010-07-22 ディフュージョン・ファーマシューティカルズ・エルエルシー 末梢血管疾患の前処置としての、及びその処置における双極性トランスカロテノイドの使用
CN108464976A (zh) 2009-06-22 2018-08-31 扩散药品有限公司 扩散促进化合物及其单独或与溶栓药一起的应用
EP2575487B1 (en) 2010-06-02 2017-10-18 Diffusion Pharmaceuticals Llc Oral formulations of bipolar trans carotenoids
WO2015023830A1 (en) 2013-08-14 2015-02-19 Stc.Unm Treatment and prevention of stroke and other neurological disorders
AU2015334455B2 (en) * 2014-10-21 2020-05-21 Gennova Biopharmaceuticals Limited A novel purification process for isolation and commercial production of recombinant TNK-tPA (Tenecteplase)
CN115089569A (zh) 2016-03-24 2022-09-23 扩散药品有限公司 双极性反式类胡萝卜素连同化疗和放射治疗在治疗癌症中的用途
CA3050858A1 (en) * 2017-01-24 2018-08-02 Ekos Corporation Method for the treatment of thromboembolism
WO2018210860A1 (en) * 2017-05-16 2018-11-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of acute ischemic stroke

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030180282A1 (en) * 2002-03-25 2003-09-25 Victor Serebruany Method of treatment of thrombotic events
US20040029798A1 (en) * 2002-02-22 2004-02-12 Paul Armstrong Combination treatment with t-PA variant and low molecular weight heparin
US20070014779A1 (en) * 2002-11-14 2007-01-18 Genentech, Inc. Plasminogen activator variant formulations

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
JP3559559B2 (ja) 1992-06-03 2004-09-02 ジェネンテク,インコーポレイテッド 向上した治療特性を有する組織プラスミノーゲン活性化因子グリコシル化変異体
JP2002173447A (ja) * 2000-09-29 2002-06-21 Yamanouchi Pharmaceut Co Ltd 脳塞栓症治療用医薬組成物
WO2006094120A2 (en) * 2005-03-02 2006-09-08 The Regents Of The University Of California Treatment for embolic stroke
ZA200900957B (en) 2006-08-29 2010-05-26 Genentech Inc Use of tenecteplase for treating acute ischemic stroke

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040029798A1 (en) * 2002-02-22 2004-02-12 Paul Armstrong Combination treatment with t-PA variant and low molecular weight heparin
US20030180282A1 (en) * 2002-03-25 2003-09-25 Victor Serebruany Method of treatment of thrombotic events
US20070014779A1 (en) * 2002-11-14 2007-01-18 Genentech, Inc. Plasminogen activator variant formulations

Cited By (65)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9023346B2 (en) 2006-08-29 2015-05-05 Genentech, Inc. Method of treating stroke with tenecteplase
US9034007B2 (en) 2007-09-21 2015-05-19 Insera Therapeutics, Inc. Distal embolic protection devices with a variable thickness microguidewire and methods for their use
US20110257510A1 (en) * 2008-12-24 2011-10-20 Koninklijke Philips Electronics N.V. System, method and apparatus for cardiac intervention with mr stroke detection and treatment
JP2013502461A (ja) * 2009-08-24 2013-01-24 ハフ イヤ インスティテュート 急性音響外傷の治療方法
US10555915B2 (en) 2009-08-24 2020-02-11 Hough Ear Institute Methods for treating acute acoustic trauma
WO2011028503A1 (en) * 2009-08-24 2011-03-10 Hough Ear Institute Methods for treating acute acoustic trauma
JP2015172071A (ja) * 2009-08-24 2015-10-01 ハフ イヤ インスティテュート 急性音響外傷の治療方法
US9943575B2 (en) * 2010-12-23 2018-04-17 Gennova Biopharmaceuticals Limited Pharmaceutical compositions of tenecteplase
US10328083B1 (en) 2012-10-18 2019-06-25 University Of South Florida Compositions and methods for treating stroke
US20150238502A1 (en) * 2012-10-18 2015-08-27 University Of South Florida Compositions and Methods for Treating Stroke
US9314324B2 (en) 2013-03-15 2016-04-19 Insera Therapeutics, Inc. Vascular treatment devices and methods
US9750524B2 (en) 2013-03-15 2017-09-05 Insera Therapeutics, Inc. Shape-set textile structure based mechanical thrombectomy systems
US11298144B2 (en) 2013-03-15 2022-04-12 Insera Therapeutics, Inc. Thrombus aspiration facilitation systems
US8733618B1 (en) 2013-03-15 2014-05-27 Insera Therapeutics, Inc. Methods of coupling parts of vascular treatment systems
US8679150B1 (en) 2013-03-15 2014-03-25 Insera Therapeutics, Inc. Shape-set textile structure based mechanical thrombectomy methods
US8747432B1 (en) 2013-03-15 2014-06-10 Insera Therapeutics, Inc. Woven vascular treatment devices
US8753371B1 (en) 2013-03-15 2014-06-17 Insera Therapeutics, Inc. Woven vascular treatment systems
US10463468B2 (en) 2013-03-15 2019-11-05 Insera Therapeutics, Inc. Thrombus aspiration with different intensity levels
US8783151B1 (en) 2013-03-15 2014-07-22 Insera Therapeutics, Inc. Methods of manufacturing vascular treatment devices
US10342655B2 (en) 2013-03-15 2019-07-09 Insera Therapeutics, Inc. Methods of treating a thrombus in an artery using cyclical aspiration patterns
US8789452B1 (en) 2013-03-15 2014-07-29 Insera Therapeutics, Inc. Methods of manufacturing woven vascular treatment devices
US10335260B2 (en) 2013-03-15 2019-07-02 Insera Therapeutics, Inc. Methods of treating a thrombus in a vein using cyclical aspiration patterns
US8690907B1 (en) 2013-03-15 2014-04-08 Insera Therapeutics, Inc. Vascular treatment methods
US10251739B2 (en) 2013-03-15 2019-04-09 Insera Therapeutics, Inc. Thrombus aspiration using an operator-selectable suction pattern
US9901435B2 (en) 2013-03-15 2018-02-27 Insera Therapeutics, Inc. Longitudinally variable vascular treatment devices
US9833251B2 (en) 2013-03-15 2017-12-05 Insera Therapeutics, Inc. Variably bulbous vascular treatment devices
US9592068B2 (en) 2013-03-15 2017-03-14 Insera Therapeutics, Inc. Free end vascular treatment systems
US9179995B2 (en) 2013-03-15 2015-11-10 Insera Therapeutics, Inc. Methods of manufacturing slotted vascular treatment devices
US8852227B1 (en) 2013-03-15 2014-10-07 Insera Therapeutics, Inc. Woven radiopaque patterns
US9179931B2 (en) 2013-03-15 2015-11-10 Insera Therapeutics, Inc. Shape-set textile structure based mechanical thrombectomy systems
US8715315B1 (en) 2013-03-15 2014-05-06 Insera Therapeutics, Inc. Vascular treatment systems
US8715314B1 (en) 2013-03-15 2014-05-06 Insera Therapeutics, Inc. Vascular treatment measurement methods
US8721677B1 (en) 2013-03-15 2014-05-13 Insera Therapeutics, Inc. Variably-shaped vascular devices
US8721676B1 (en) 2013-03-15 2014-05-13 Insera Therapeutics, Inc. Slotted vascular treatment devices
US8910555B2 (en) 2013-03-15 2014-12-16 Insera Therapeutics, Inc. Non-cylindrical mandrels
US8904914B2 (en) 2013-03-15 2014-12-09 Insera Therapeutics, Inc. Methods of using non-cylindrical mandrels
US8882797B2 (en) 2013-03-15 2014-11-11 Insera Therapeutics, Inc. Methods of embolic filtering
US8895891B2 (en) 2013-03-15 2014-11-25 Insera Therapeutics, Inc. Methods of cutting tubular devices
US8728117B1 (en) 2013-07-29 2014-05-20 Insera Therapeutics, Inc. Flow disrupting devices
US8728116B1 (en) 2013-07-29 2014-05-20 Insera Therapeutics, Inc. Slotted catheters
US8870910B1 (en) 2013-07-29 2014-10-28 Insera Therapeutics, Inc. Methods of decoupling joints
US8932321B1 (en) 2013-07-29 2015-01-13 Insera Therapeutics, Inc. Aspiration systems
US8872068B1 (en) 2013-07-29 2014-10-28 Insera Therapeutics, Inc. Devices for modifying hypotubes
US8869670B1 (en) 2013-07-29 2014-10-28 Insera Therapeutics, Inc. Methods of manufacturing variable porosity devices
US8863631B1 (en) 2013-07-29 2014-10-21 Insera Therapeutics, Inc. Methods of manufacturing flow diverting devices
US8866049B1 (en) 2013-07-29 2014-10-21 Insera Therapeutics, Inc. Methods of selectively heat treating tubular devices
US8859934B1 (en) 2013-07-29 2014-10-14 Insera Therapeutics, Inc. Methods for slag removal
US8845678B1 (en) 2013-07-29 2014-09-30 Insera Therapeutics Inc. Two-way shape memory vascular treatment methods
US8715316B1 (en) 2013-07-29 2014-05-06 Insera Therapeutics, Inc. Offset vascular treatment devices
US8845679B1 (en) 2013-07-29 2014-09-30 Insera Therapeutics, Inc. Variable porosity flow diverting devices
US8932320B1 (en) 2013-07-29 2015-01-13 Insera Therapeutics, Inc. Methods of aspirating thrombi
US8870901B1 (en) 2013-07-29 2014-10-28 Insera Therapeutics, Inc. Two-way shape memory vascular treatment systems
US8828045B1 (en) 2013-07-29 2014-09-09 Insera Therapeutics, Inc. Balloon catheters
US8715317B1 (en) 2013-07-29 2014-05-06 Insera Therapeutics, Inc. Flow diverting devices
US8813625B1 (en) 2013-07-29 2014-08-26 Insera Therapeutics, Inc. Methods of manufacturing variable porosity flow diverting devices
US8803030B1 (en) 2013-07-29 2014-08-12 Insera Therapeutics, Inc. Devices for slag removal
US8795330B1 (en) 2013-07-29 2014-08-05 Insera Therapeutics, Inc. Fistula flow disruptors
US8790365B1 (en) 2013-07-29 2014-07-29 Insera Therapeutics, Inc. Fistula flow disruptor methods
US10390926B2 (en) 2013-07-29 2019-08-27 Insera Therapeutics, Inc. Aspiration devices and methods
US8816247B1 (en) 2013-07-29 2014-08-26 Insera Therapeutics, Inc. Methods for modifying hypotubes
US8784446B1 (en) 2013-07-29 2014-07-22 Insera Therapeutics, Inc. Circumferentially offset variable porosity devices
US8735777B1 (en) 2013-07-29 2014-05-27 Insera Therapeutics, Inc. Heat treatment systems
US10751159B2 (en) 2013-07-29 2020-08-25 Insera Therapeutics, Inc. Systems for aspirating thrombus during neurosurgical procedures
US10426424B2 (en) 2017-11-21 2019-10-01 General Electric Company System and method for generating and performing imaging protocol simulations
WO2021158799A1 (en) * 2020-02-04 2021-08-12 The Regents Of The University Of Colorado, A Body Corporate Prophylactic uses of annexin a2

Also Published As

Publication number Publication date
SG10201804944XA (en) 2018-07-30
JP5785687B2 (ja) 2015-09-30
CN101505785A (zh) 2009-08-12
EP2056856A2 (en) 2009-05-13
BRPI0716143A2 (pt) 2013-11-26
UA97486C2 (ru) 2012-02-27
ZA200900957B (en) 2010-05-26
HK1131054A1 (en) 2010-01-15
EA200970237A1 (ru) 2009-08-28
WO2008027687A2 (en) 2008-03-06
MY163119A (en) 2017-08-15
US20120164157A1 (en) 2012-06-28
IL196987A0 (en) 2011-08-01
CA2661012C (en) 2019-03-19
SG174753A1 (en) 2011-10-28
CA2661012A1 (en) 2008-03-06
IL248176A0 (en) 2016-11-30
ECSP099178A (es) 2009-06-30
IL196987A (en) 2016-10-31
AU2007290277B2 (en) 2013-04-11
EA015573B1 (ru) 2011-10-31
NO342302B1 (no) 2018-04-30
US9023346B2 (en) 2015-05-05
EP2056856B1 (en) 2015-01-14
JP2010502629A (ja) 2010-01-28
CO6150188A2 (es) 2010-04-20
TWI439282B (zh) 2014-06-01
WO2008027687A3 (en) 2008-05-22
KR20090045405A (ko) 2009-05-07
NO20091296L (no) 2009-05-28
NZ574767A (en) 2012-02-24
AU2007290277A1 (en) 2008-03-06
TW200817033A (en) 2008-04-16
MX2009001918A (es) 2009-03-06
SG10201501498VA (en) 2015-04-29

Similar Documents

Publication Publication Date Title
US9023346B2 (en) Method of treating stroke with tenecteplase
Barreto Intravenous thrombolytics for ischemic stroke
Furlan et al. Thrombolytic therapy in acute ischemic stroke
US20200255530A1 (en) Compositions and methods for treatment of stroke and other cns disorders
EP4245313A1 (en) Extended, high dose vegf antagonist regimens for treatment of angiogenic eye disorders
Broderick Recanalization therapies for acute ischemic stroke
US20220073888A1 (en) Combination treatment
Movva et al. There is no difference in safety and efficacy with tirofiban or eptifibatide for patients undergoing treatment of large vessel occlusion and underlying intracranial atherosclerosis
EP2289542A1 (en) Treatment of neurological or neurodegenerative disorders
Brott Reopening occluded cerebral arteries
EP4245312A1 (en) Extended, high dose vegf antagonist regimens for treatment of angiogenic eye disorders
Marè et al. Comparison of pharmacokinetic properties of alteplase and tenecteplase. The future of thrombolysis in acute ischemic stroke
Weiner Thrombolytic agents in critical care
Spinler et al. Update on strategies to improve thrombolysis for acute myocardial infarction
KR20210014024A (ko) 트리사이클릭 유도체를 이용한 뇌졸중의 치료 방법
Mishira Antithrombotic therapy: current status and future developments
Hoang et al. The efficacy and safety of tissue plasminogen activator in acute ischemic strokes
CN117295497A (zh) 用于治疗脑梗塞的药物组合物
KR100831770B1 (ko) t-PA 변이체와 저분자량 헤파린의 조합 치료
JP2008507561A (ja) 活性化プロテインcによる、脳内のプラスミノゲン活性化因子の不都合な作用の阻害
Zhang et al. Safety of tirofiban for patients with acute ischemic stroke in routine clinical practice
WO2007134813A2 (en) Use of desmodus salivary plasminogen activator (dspa) for treating venous thromboembolism
Diaz-Ricart Desmoteplase
Frishman et al. Antithrombotic therapy
EP2289541A1 (en) Treatment of neurological or neurodegenerative disorders

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENENTECH, INC, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KUEBLER, PETER;REEL/FRAME:020406/0382

Effective date: 20080111

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION