1
|
Li S, Gu HQ, Dai H, Lu G, Wang Y. Reteplase versus alteplase for acute ischaemic stroke within 4.5 hours (RAISE): rationale and design of a multicentre, prospective, randomised, open-label, blinded-endpoint, controlled phase 3 non-inferiority trial. Stroke Vasc Neurol 2024; 9:568-573. [PMID: 38286482 DOI: 10.1136/svn-2023-003035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 12/23/2023] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND AND PURPOSE Reteplase is the third generation of alternative thrombolytic agent. We hypothesis that reteplase will be non-inferior to alteplase in achieving excellent functional outcome at 90 days among eligible patients with acute ischaemic stroke. METHODS AND DESIGN Reteplase versus alteplase for acute ischaemic stroke within 4.5 hours (RAISE) trial is a multicentre, prospective, randomised, open-label, blinded endpoint (PROBE), controlled phase 3 non-inferiority trial. A total of 1412 eligible patients will be randomly assigned to receive either reteplase at a dose of 18 mg+ 18 mg or alteplase 0.9 mg/kg at a ratio of 1:1. An independent data monitoring committee will review the trail's progress and safety data. STUDY OUTCOMES The primary efficacy outcome of this study is proportion of individuals attaining an excellent functional outcome, defined as modified Rankin Scale (mRS) 0-1 at 90 days. The secondary efficacy outcomes encompass favourable functional outcome defined as mRS 0-2, major neurological improvement on the National Institutes of Health Stroke Scale, ordinal distribution of mRS and Barthel Index score of at least 95 points at 90 days. The primary safety outcomes are symptomatic intracranial haemorrhage at 36 hours within 90 days. DISCUSSION The RAISE trial will provide crucial insights into the selection of thrombolytic agents for stroke thrombolysis. TRIAL REGISTRATION NUMBER NCT05295173.
Collapse
Affiliation(s)
- Shuya Li
- Department of Neurology, and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hong-Qiu Gu
- Department of Neurology, and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hongguo Dai
- Department of Emergency, Linfen Central Hospital, Shanxi Province, China
| | - Guozhi Lu
- Department of Neurology, Keshiketeng Banner Traditional Chinese Medicine Mongolian Medical Hospital, The Inner Mongolia autonomous region, China
| | - Yongjun Wang
- Department of Neurology, and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Li S, Wang X, Jin A, Liu G, Gu H, Li H, Campbell BCV, Fisher M, Yang Y, Wei Y, Wang J, Wang Y, Zhao X, Liu L, Li Z, Meng X, Wang Y. Safety and Efficacy of Reteplase Versus Alteplase for Acute Ischemic Stroke: A Phase 2 Randomized Controlled Trial. Stroke 2024; 55:366-375. [PMID: 38152962 DOI: 10.1161/strokeaha.123.045193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/05/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Reteplase is a more affordable new-generation thrombolytic with a prolonged half-life. We aimed to determine the safety dose range of reteplase for patients with acute ischemic stroke within 4.5 hours of onset. METHODS This is a multicenter, prospective, randomized controlled, open-label, blinded-end point phase 2 clinical trial. Patients with acute ischemic stroke aged between 18 and 80 years who were eligible for standard intravenous thrombolysis were enrolled from 17 centers in China and randomly assigned (1:1:1) to receive intravenous reteplase 12+12 mg, intravenous reteplase 18+18 mg, or intravenous alteplase 0.9 mg/kg. The primary safety outcome was symptomatic intracranial hemorrhage (SITS definition) within 36 hours. The primary efficacy outcome was the proportion of patients with the National Institutes of Health Stroke Scale score of no more than 1 or a decrease of at least 4 points from the baseline at 14 days after thrombolysis. RESULTS Between August 2019 and May 2021, 180 patients were randomly assigned to reteplase 12+12 mg (n=61), reteplase 18+18 mg (n=67), or alteplase (n=52). Four patients did not receive the study agent. Symptomatic intracranial hemorrhage occurred in 3 of 60 (5.0%) in the reteplase 12+12 mg group, 1 of 66 (1.5%) in the reteplase 18+18 mg group, and 1 of 50 (2.0%) in the alteplase group (P=0.53). The primary efficacy outcome in the modified intention-to-treat population occurred in 45 of 60 (75.0%) in the reteplase 12+12 mg group (odds ratio, 0.85 [95% CI, 0.35-2.06]), 48 of 66 (72.7%) in the reteplase 18+18 mg group (odds ratio, 0.75 [95% CI, 0.32-1.78]), and 39 of 50 (78.0%) in alteplase group. CONCLUSIONS Reteplase was well tolerated in patients with acute ischemic stroke within 4.5 hours of onset in China with a similar efficacy profile to alteplase. The efficacy and appropriate dosage of reteplase for patients with acute ischemic stroke need prospective validation. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT04028518.
Collapse
Affiliation(s)
- Shuya Li
- Department of Neurology and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
| | - Xuechun Wang
- Department of Neurology and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
| | - Aoming Jin
- Department of Neurology and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
| | - Gaifen Liu
- Department of Neurology and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
| | - Hongqiu Gu
- Department of Neurology and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
| | - Hao Li
- Department of Neurology and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
| | - Bruce C V Campbell
- Department of Medicine and Neurology, Melbourne Brain Centre at the Royal Melbourne Hospital, The University of Melbourne, VIC, Australia (B.C.V.C.)
| | - Marc Fisher
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (M.F.)
| | - Yi Yang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China (Y.Y.)
| | - Yan Wei
- Department of Neurology, Halison International Peace Hospital of Hengshui City, China (Y.W.)
| | - Junhai Wang
- Department of Neurology, Sinopharm Tongmei General Hospital, Datong, China (J.W.)
| | - Yilong Wang
- Department of Neurology and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
| | - Xingquan Zhao
- Department of Neurology and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
| | - Liping Liu
- Department of Neurology and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
| | - Zixiao Li
- Department of Neurology and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
| | - Xia Meng
- Department of Neurology and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
| | - Yongjun Wang
- Department of Neurology and Department of Clinical Trial Center, Beijing Tiantan Hospital, Capital Medical University, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.L., X.W., A.J., G.L., H.G., H.L., Yilong Wang, X.Z., L.L., Z.L., X.M., Yongjun Wang)
| |
Collapse
|
3
|
Izadi S, Jalali Javaran M, Rashidi Monfared S, Castilho A. Reteplase Fc-fusions produced in N. benthamiana are able to dissolve blood clots ex vivo. PLoS One 2021; 16:e0260796. [PMID: 34847186 PMCID: PMC8631678 DOI: 10.1371/journal.pone.0260796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/16/2021] [Indexed: 11/19/2022] Open
Abstract
Thrombolytic and fibrinolytic therapies are effective treatments to dissolve blood clots in stroke therapy. Thrombolytic drugs activate plasminogen to its cleaved form plasmin, a proteolytic enzyme that breaks the crosslinks between fibrin molecules. The FDA-approved human tissue plasminogen activator Reteplase (rPA) is a non-glycosylated protein produced in E. coli. rPA is a deletion mutant of the wild-type Alteplase that benefits from an extended plasma half-life, reduced fibrin specificity and the ability to better penetrate into blood clots. Different methods have been proposed to improve the production of rPA. Here we show for the first time the transient expression in Nicotiana benthamiana of rPA fused to the immunoglobulin fragment crystallizable (Fc) domain on an IgG1, a strategy commonly used to improve the stability of therapeutic proteins. Despite our success on the expression and purification of dimeric rPA-Fc fusions, protein instability results in high amounts of Fc-derived degradation products. We hypothesize that the "Y"- shape of dimeric Fc fusions cause steric hindrance between protein domains and leads to physical instability. Indeed, mutations of critical residues in the Fc dimerization interface allowed the expression of fully stable rPA monomeric Fc-fusions. The ability of rPA-Fc to convert plasminogen into plasmin was demonstrated by plasminogen zymography and clot lysis assay shows that rPA-Fc is able to dissolve blood clots ex vivo. Finally, we addressed concerns with the plant-specific glycosylation by modulating rPA-Fc glycosylation towards serum-like structures including α2,6-sialylated and α1,6-core fucosylated N-glycans completely devoid of plant core fucose and xylose residues.
Collapse
Affiliation(s)
- Shiva Izadi
- Department of Applied Genetics and Cell Biology, Natural Resources and Life Sciences, Vienna, Austria
- Faculty of Agriculture, Department of Plant Genetics and Breeding, Tarbiat Modares University, Tehran, Iran
| | - Mokhtar Jalali Javaran
- Faculty of Agriculture, Department of Agricultural Biotechnology, Tarbiat Modares University, Tehran, Iran
| | - Sajad Rashidi Monfared
- Faculty of Agriculture, Department of Agricultural Biotechnology, Tarbiat Modares University, Tehran, Iran
| | - Alexandra Castilho
- Department of Applied Genetics and Cell Biology, Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
4
|
Abstract
Generation of plasmin in the vicinity of a blood clot has proven to be an effective approach for treating thrombotic disorders, particularly myocardial infarction. Conceptually, the ideal thrombolytic agent would initiate the formation of plasmin, primarily in association with fibrin incorporated into the occlusive thrombus. Thus, thrombolytic agents that exhibit relative fibrin specificity and, thus, presumably clot selectivity (e.g., tissue plasminogen activator) were expected to have a marked clinical benefit compared to agents that do not display affinity for fibrin (e.g., streptokinase). However, results obtained recently from clinical trials indicate that these 2 agents essentially were equally effective in treating myocardial infarction. With these findings in mind, efforts are being made to develop novel thrombolytic agents that might achieve more rapid and specific thrombolysis than that achieved by presently available agents and, thus, could be administered earlier because of an improved margin of safety. The available data suggest that tissue-type PA (tPA) mutants possessing resistance to endogenous inhibitors, altered fibrin affinity, and/or slower rates of clearance may prove beneficial in this regard. In addition, adjunctive therapies (i.e., anti-platelet and anti-thrombin compounds) have been found to decrease the time necessary to achieve reperfusion and have reduced rates of reocclusion. These efforts are expected to yield therapeutic agents in the 1990s and beyond that, when administered in combination, would exhibit increased efficacy in the treatment of myocardial infarction and other thrombotic disorders.
Collapse
Affiliation(s)
- Laurence A. Erickson
- Cardiovascular Diseases and Molecular Biology Research, Upjohn Laboratories, Kalamazoo, Michigan
| | - Keith R. Marotti
- Cardiovascular Diseases and Molecular Biology Research, Upjohn Laboratories, Kalamazoo, Michigan
| |
Collapse
|
5
|
Nedaeinia R, Faraji H, Javanmard SH, Ferns GA, Ghayour-Mobarhan M, Goli M, Mashkani B, Nedaeinia M, Haghighi MHH, Ranjbar M. Bacterial staphylokinase as a promising third-generation drug in the treatment for vascular occlusion. Mol Biol Rep 2019; 47:819-841. [PMID: 31677034 DOI: 10.1007/s11033-019-05167-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022]
Abstract
Vascular occlusion is one of the major causes of mortality and morbidity. Blood vessel blockage can lead to thrombotic complications such as myocardial infarction, stroke, deep venous thrombosis, peripheral occlusive disease, and pulmonary embolism. Thrombolytic therapy currently aims to rectify this through the administration of recombinant tissue plasminogen activator. Research is underway to design an ideal thrombolytic drug with the lowest risk. Despite the potent clot lysis achievable using approved thrombolytic drugs such as alteplase, reteplase, streptokinase, tenecteplase, and some other fibrinolytic agents, there are some drawbacks, such as high production cost, systemic bleeding, intracranial hemorrhage, vessel re-occlusion by platelet-rich and retracted secondary clots, and non-fibrin specificity. In comparison, bacterial staphylokinase, is a new, small-size plasminogen activator, unlike bacterial streptokinase, it hinders the systemic degradation of fibrinogen and reduces the risk of severe hemorrhage. A fibrin-bound plasmin-staphylokinase complex shows high resistance to a2-antiplasmin-related inhibition. Staphylokinase has the potential to be considered as a promising thrombolytic agent with properties of cost-effective production and the least side effects.
Collapse
Affiliation(s)
- Reza Nedaeinia
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Habibollah Faraji
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran. .,Department of Laboratory Sciences, Faculty of Para-Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Shaghayegh Haghjooye Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Science, Isfahan, Iran
| | - Gordon A Ferns
- Brighton and Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, BN1 9PH, UK
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Goli
- Department of Food Science and Technology, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Baratali Mashkani
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mozhdeh Nedaeinia
- Young Researchers and Elite Club, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Mohammad Hossein Hayavi Haghighi
- Department of Health Information Management, Faculty of Para-Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Maryam Ranjbar
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran.,Deputy of Food and Drug, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
6
|
Salama Y, Lin SY, Dhahri D, Hattori K, Heissig B. The fibrinolytic factor tPA drives LRP1-mediated melanoma growth and metastasis. FASEB J 2018; 33:3465-3480. [PMID: 30458112 DOI: 10.1096/fj.201801339rrr] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The multifunctional endocytic receptor low-density lipoprotein receptor-related protein (LRP)1 has recently been identified as a hub within a biomarker network for multicancer clinical outcome prediction. The mechanism how LRP1 modulates cancer progression is poorly understood. In this study we found that LRP1 and one of its ligands, tissue plasminogen activator (tPA), are expressed in melanoma cells and control melanoma growth and lung metastasis in vivo. Mechanistic studies were performed on 2 melanoma cancer cell lines, B16F10 and the B16F1 cells, both of which form primary melanoma tumors, but only B16F10 cells metastasize to the lungs. Tumor-, but not niche cell-derived tPA, enhanced melanoma cell proliferation in tPA-/- mice. Gain-of-function experiments revealed that melanoma LRP1 is critical for tumor growth, recruitment of mesenchymal stem cells into the tumor bed, and metastasis. Melanoma LRP1 was found to enhance ERK activation, resulting in increased matrix metalloproteinase (MMP)-9 RNA, protein, and secreted activity, a well-known modulator of melanoma metastasis. Restoration of LRP1 and tPA in the less aggressive, poorly metastatic B16F1 tumor cells enhanced tumor cell proliferation and led to massive lung metastasis in murine tumor models. Antimelanoma drug treatment induced tPA and LRP1 expression. tPA or LRP1 knockdown enhanced chemosensitivity in melanoma cells. Our results identify the tPA-LRP1 pathway as a key switch that drives melanoma progression, in part by modulating the cellular composition and proteolytic makeup of the tumor niche. Targeting this pathway may be a novel treatment strategy in combination treatments for melanoma.-Salama, Y., Lin, S.-Y., Dhahri, D., Hattori, K., Heissig, B. The fibrinolytic factor tPA drives LRP1-mediated melanoma growth and metastasis.
Collapse
Affiliation(s)
- Yousef Salama
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Shiou-Yuh Lin
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Douaa Dhahri
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koichi Hattori
- Center for Genome and Regenerative Medicine Juntendo University School of Medicine, Tokyo, Japan; and
| | - Beate Heissig
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Atopy (Allergy) Center, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
7
|
Can the benefits of rtPA treatment for stroke be improved? Rev Neurol (Paris) 2017; 173:566-571. [PMID: 28797689 DOI: 10.1016/j.neurol.2017.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/02/2017] [Accepted: 07/07/2017] [Indexed: 12/14/2022]
Abstract
Tissue-type plasminogen activator (tPA) is a serine protease well known to promote fibrinolysis. This is why: its recombinant form (rtPA) can be used, either alone or combined with thrombectomy, to promote recanalization/reperfusion following ischemic stroke. However, its overall benefits are counteracted by some of its side-effects, including incomplete lysis of clots, an increased risk of hemorrhagic transformation and the possibility of neurotoxicity. Nevertheless, better understanding of the mechanisms by which tPA influences brain function and promotes its alteration may help in the design of new strategies to improve stroke therapy.
Collapse
|
8
|
Scale up and pharmacokinetic study of a novel mutated chimeric tissue plasminogen activator (mt-PA) in rats. Sci Rep 2017; 7:43028. [PMID: 28223717 PMCID: PMC5320447 DOI: 10.1038/srep43028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/17/2017] [Indexed: 11/08/2022] Open
Abstract
Because of high mortality caused by cardiovascular diseases, various fibrinolytic agents with diverse pharmacokinetic and pharmacodynamic properties have been developed. A novel mutated chimeric tissue plasminogen activator (mt-PA) was developed by the removal of first three domains of t-PA, insertion of GHRP sequence and mutation towards resistance to plasminogen activator inhibitor-1 (PAI-1). Mt-PA protein was expressed in Expi293F cells. The expression level of mt-PA was found to be 5000 IU/mL. Following purification, the pharmacokinetic properties of mt-PA were evaluated in three doses in rats. Data related to mt-PA were best fitted to two compartment model. With the increase in dose, the Area Under the plasma concentration-time Curve (AUC0→∞) increased. The elimination half-life (t1/2) of mt-PA was in the range of 19.1–26.1 min in three doses while that of Alteplase was 8.3 min. The plasma clearance (CLp) of mt-PA ranged from 3.8 to 5.9 mL/min in three doses, which was several times lower than that of Alteplase (142.6 mL/min). The mean residence time (MRT) of mt-PA ranged from 23.3–31.8 min in three doses, which was 4–5 times greater than that of Alteplase (6 min). Mt-PA showed extended half-life and mean residence time and is a good candidate for further clinical studies.
Collapse
|
9
|
Chevilley A, Lesept F, Lenoir S, Ali C, Parcq J, Vivien D. Impacts of tissue-type plasminogen activator (tPA) on neuronal survival. Front Cell Neurosci 2015; 9:415. [PMID: 26528141 PMCID: PMC4607783 DOI: 10.3389/fncel.2015.00415] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 10/01/2015] [Indexed: 11/18/2022] Open
Abstract
Tissue-type plasminogen activator (tPA) a serine protease is constituted of five functional domains through which it interacts with different substrates, binding proteins, and receptors. In the last years, great interest has been given to the clinical relevance of targeting tPA in different diseases of the central nervous system, in particular stroke. Among its reported functions in the central nervous system, tPA displays both neurotrophic and neurotoxic effects. How can the protease mediate such opposite functions remain unclear but several hypotheses have been proposed. These include an influence of the degree of maturity and/or the type of neurons, of the level of tPA, of its origin (endogenous or exogenous) or of its form (single chain tPA versus two chain tPA). In this review, we will provide a synthetic snapshot of our current knowledge regarding the natural history of tPA and discuss how it sustains its pleiotropic functions with focus on excitotoxic/ischemic neuronal death and neuronal survival.
Collapse
Affiliation(s)
- Arnaud Chevilley
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Flavie Lesept
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Sophie Lenoir
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Carine Ali
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Jérôme Parcq
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Denis Vivien
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| |
Collapse
|
10
|
Docagne F, Parcq J, Lijnen R, Ali C, Vivien D. Understanding the Functions of Endogenous and Exogenous Tissue-Type Plasminogen Activator During Stroke. Stroke 2015; 46:314-20. [DOI: 10.1161/strokeaha.114.006698] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Fabian Docagne
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| | - Jérôme Parcq
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| | - Roger Lijnen
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| | - Carine Ali
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| | - Denis Vivien
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| |
Collapse
|
11
|
Cloning and expression of hybrid streptokinase towards clot-specific activity. J Microbiol Methods 2014; 98:84-8. [PMID: 24440164 DOI: 10.1016/j.mimet.2014.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 01/08/2014] [Accepted: 01/09/2014] [Indexed: 11/21/2022]
Abstract
Streptokinase (SK) is a thrombolytic agent that is widely used to treat myocardial infarction and pulmonary embolism. The lack of fibrin specificity of SK for the clot lysis is one of the limitations of SK. In this study, we have incorporated the finger and Kringle 2 domains from the human tissue type plasminogen activator gene (t-PA) at the 5' end of the SK gene. These domains are responsible for specific binding to fibrin. We have used the pRSETB vector in an attempt to express the hybrid streptokinase possessing specificity for fibrin. On this regard, three hybrid streptokinase were constructed and expressed in Escherichia coli BL21 (DE3): the finger domain with SK (FSK), the Kringle 2 domain with SK (KSK) and the finger domain+Kringle 2 with SK (FKSK). The activities of the hybrid SKs were assessed by caseinolytic assay and clot lysis assay. All hybrid SKs were found to activate plasminogen in the caseinolytic plate assay. In the clot lysis assay, KSK and FSK were able to dissolute human blood and artificial clots in a fibrin-dependent manner unlike the SK and FKSK proteins.
Collapse
|
12
|
Lee SB, Oh HK, Kim HK, Joe YA. Expression of the non-glycosylated kringle domain of tissue type plasminogen activator in Pichia and its anti-endothelial cell activity. Protein Expr Purif 2006; 50:1-8. [PMID: 16854593 DOI: 10.1016/j.pep.2006.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2006] [Revised: 05/30/2006] [Accepted: 06/01/2006] [Indexed: 10/24/2022]
Abstract
The two-kringle domain of tissue-type plasminogen activator (TK1-2) has been identified as a potent angiogenesis inhibitor by suppressing endothelial cell proliferation, in vivo angiogenesis, and in vivo tumor growth. Escherichia coli-derived, non-glycosylated TK1-2 more potently inhibits in vivo tumor growth, whereas Pichia expression system is more efficient for producing TK1-2 as a soluble form, albeit accompanying N-glycosylation. Therefore, in order to avoid immune reactivity and improve in vivo efficacy, we expressed the non-glycosylated form of TK1-2 in Pichia pastoris and evaluated its activity in vitro. When TK1-2 was mutated at either Asn(117) or Asn(184) by replacing with Gln, the mutated proteins produced the glycosylated form in Pichia, of which sugar moiety could be deleted by endoglycosidase H treatment. When both sites were replaced by Gln, the resulting mutant produced a non-glycosylated protein, NQ-TK1-2. Secreted NQ-TK1-2 was purified from the culture broth by sequential ion exchange chromatography using SP-sepharose, Q-spin, and UNO-S1 column. The purified NQ-TK1-2 migrated as a single protein band of approximately 20 kDa in SDS-PAGE and its mass spectrum showed one major peak of 19,950.71 Da, which is smaller than those of two glycosylated forms of wild type TK1-2. Functionally, the purified NQ-TK1-2 inhibited endothelial cell proliferation and migration stimulated by bFGF and VEGF, respectively. Therefore, the results suggest that non-glycosylated TK1-2 useful for the treatment of cancer can be efficiently produced in Pichia, with retaining its activity.
Collapse
Affiliation(s)
- Sang-Bae Lee
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | | | | | | |
Collapse
|
13
|
Thrombolysis in children with congenital heart disease. PROGRESS IN PEDIATRIC CARDIOLOGY 2005. [DOI: 10.1016/j.ppedcard.2005.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
14
|
Fukao H, Ueshima S, Okada K, Matsuo O. Binding of mutant tissue-type plasminogen activators to human endothelial cells and their extracellular matrix. Life Sci 2000; 66:2473-87. [PMID: 10894090 DOI: 10.1016/s0024-3205(00)80007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We previously demonstrated that tissue-type plasminogen activator (t-PA) specifically bound to its receptor (t-PAR) on human umbilical vein endothelial cells (HUVEC). In addition to analyses of t-PA binding to plasminogen activator inhibitor-1 (PAI-1) in the extracellular matrix (ECM) and to the t-PAR, we further evaluated the binding of three t-PA mutants, deltaFE1X t-PA lacking finger (F), epidermal growth factor-like (E) domains and one sugar chain at Asn177 thus comprising two kringles (K1 and K2) and protease (P) domains, deltaFE3X t-PA with three glycosylation sites deleted at Asn117, 184, and 448, and deltaFEK1 t-PA comprising K2 and P domains without glycosylation. Wild-type t-PA bound to ECM with high affinity, which was completely blocked by anti-PAI-1 IgG. Wild-type t-PA, deltaFE1X t-PA and deltaFEK1 t-PA bound to two classes of binding sites with high and low affinities on monolayer HUVEC. However, all t-PAs bound to a single class of binding site in the presence of anti-PAI-1 IgG. DeltaFEK1 t-PA bound t-PAR maximally among these t-PAs. These results suggested that the high affinity binding of t-PA mainly occurred with PAI-1 on ECM while the low affinity binding was with t-PAR. The deletion of F, E domains and sugar chains had no effect on binding with t-PAR. However, since only K1-missing t-PA (deltaFEK1) exhibited significantly increased binding sites among these t-PAs, it was suggested that the binding to t-PAR was mediated mainly by K2 domain and that the increase of binding was due to direct exposure of K2 domain.
Collapse
Affiliation(s)
- H Fukao
- Department of Physiology, Kinki University School of Medicine, Osakasayama city, Osaka, Japan.
| | | | | | | |
Collapse
|
15
|
Podor TJ, Peterson CB, Lawrence DA, Stefansson S, Shaughnessy SG, Foulon DM, Butcher M, Weitz JI. Type 1 plasminogen activator inhibitor binds to fibrin via vitronectin. J Biol Chem 2000; 275:19788-94. [PMID: 10764803 DOI: 10.1074/jbc.m908079199] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Type 1 plasminogen activator inhibitor (PAI-1), the primary inhibitor of tissue-type plasminogen activator (t-PA), circulates as a complex with the abundant plasma glycoprotein, vitronectin. This interaction stabilizes the inhibitor in its active conformation In this report, the effects of vitronectin on the interactions of PAI-1 with fibrin clots were studied. Confocal microscopic imaging of platelet-poor plasma clots reveals that essentially all fibrin-associated PAI-1 colocalizes with fibrin-bound vitronectin. Moreover, formation of platelet-poor plasma clots in the presence of polyclonal antibodies specific for vitronectin attenuated the inhibitory effects of PAI-1 on t-PA-mediated fibrinolysis. Addition of vitronectin during clot formation markedly potentiates PAI-1-mediated inhibition of lysis of (125)I-labeled fibrin clots by t-PA. This effect is dependent on direct binding interactions of vitronectin with fibrin. There is no significant effect of fibrin-associated vitronectin on fibrinolysis in the absence of PAI-1. The binding of PAI-1 to fibrin clots formed in the absence of vitronectin was characterized by a low affinity (K(d) approximately 3.5 micrometer) and rapid loss of PAI-1 inhibitory activity over time. In contrast, a high affinity and stabilization of PAI-1 activity characterized the cooperative binding of PAI-1 to fibrin formed in the presence of vitronectin. These findings indicate that plasma PAI-1.vitronectin complexes can be localized to the surface of fibrin clots; by this localization, they may modulate fibrinolysis and clot reorganization.
Collapse
Affiliation(s)
- T J Podor
- Department of Pathology and Molecular Medicine, McMaster University and the Hamilton Civic Hospitals Research Centre, Hamilton, Ontario L8V 1C3, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
Tissue plasminogen activator (tPA) is a thrombolytic agent that activates plasminogen into plasmin almost exclusively in the presence of fibrin. Intraocular injection of tPA has been proposed for the treatment of vitreoretinal diseases, such as vitreous hemorrhage, postvitrectomy fibrin formation, submacular hemorrhage, retinal vascular occlusive disorders, suprachoroidal hemorrhage and endophthalmitis. Currently, intraocular tPA is only used in the treatment of postvitrectomy fibrin formation and submacular hemorrhage. For other indications, tPA has not been shown to be safe or effective. This article reviews the use of tPA in the treatment of vitreoretinal disorders.
Collapse
Affiliation(s)
- M Kamei
- Department of Ophthalmology, Osaka University Medical School, Suita, Japan.
| | | | | |
Collapse
|
17
|
Abstract
Ultrasound reversibly alters the structure of polymerized fibrin, an effect that could influence tissue-plasminogen activator (t-PA) binding. We have, therefore, characterized the effects of ultrasound on binding of t-PA to fibrin using a novel system in which radiolabeled, active-site blocked, single chain tissue-plasminogen activator flowed through a fibrin gel at constant rate, and specific binding was determined by monitoring incorporation of radiolabel. Results using polymerized fibrin were compared with those using a surface of fibrin immobilized on Sepharose beads in a similar system. Interaction of t-PA with surface-immobilized fibrin involved two classes of binding sites (Kd = 31 nmol/L and 244 nmol/L) and a maximum binding ratio of 3.8 mol t-PA/mol fibrin. Ultrasound increased Kd for the high affinity site to 46 nmol/L (P < .0001), but it had no significant effects on the Kd 244 nmol/L site nor on Bmax. Tissue-plasminogen activator binding to noncrosslinked fibrin involved two sites with Kds of 267 nmol/L and 952 nmol/L, while a single Kd 405 nmol/L site was identified for crosslinked fibrin. Ultrasound had no significant effect on the binding affinity for noncrosslinked fibrin, but Bmaxwas increased in the presence of ultrasound, from 31 μmol/L to 43 μmol/L (P < .0001). Ultrasound decreased the Kd for crosslinked fibrin to 343 nmol/L (P = .026) and also increased Bmax from 22 μmol/L to 25 μmol/L (P = .015). Ultrasound also affected the kinetics of t-PA binding to fibrin, significantly accelerating the rate of dissociation by 77% ± 5% for noncrosslinked fibrin and by 69% ± 3% for crosslinked fibrin (P < .001 for each). These results indicate that ultrasound exposure accelerates t-PA binding, alters binding affinity, and increases maximum binding to polymerized fibrin, effects that may result from ultrasound-induced changes in fibrin structure.
Collapse
|
18
|
Abstract
AbstractUltrasound reversibly alters the structure of polymerized fibrin, an effect that could influence tissue-plasminogen activator (t-PA) binding. We have, therefore, characterized the effects of ultrasound on binding of t-PA to fibrin using a novel system in which radiolabeled, active-site blocked, single chain tissue-plasminogen activator flowed through a fibrin gel at constant rate, and specific binding was determined by monitoring incorporation of radiolabel. Results using polymerized fibrin were compared with those using a surface of fibrin immobilized on Sepharose beads in a similar system. Interaction of t-PA with surface-immobilized fibrin involved two classes of binding sites (Kd = 31 nmol/L and 244 nmol/L) and a maximum binding ratio of 3.8 mol t-PA/mol fibrin. Ultrasound increased Kd for the high affinity site to 46 nmol/L (P < .0001), but it had no significant effects on the Kd 244 nmol/L site nor on Bmax. Tissue-plasminogen activator binding to noncrosslinked fibrin involved two sites with Kds of 267 nmol/L and 952 nmol/L, while a single Kd 405 nmol/L site was identified for crosslinked fibrin. Ultrasound had no significant effect on the binding affinity for noncrosslinked fibrin, but Bmaxwas increased in the presence of ultrasound, from 31 μmol/L to 43 μmol/L (P < .0001). Ultrasound decreased the Kd for crosslinked fibrin to 343 nmol/L (P = .026) and also increased Bmax from 22 μmol/L to 25 μmol/L (P = .015). Ultrasound also affected the kinetics of t-PA binding to fibrin, significantly accelerating the rate of dissociation by 77% ± 5% for noncrosslinked fibrin and by 69% ± 3% for crosslinked fibrin (P < .001 for each). These results indicate that ultrasound exposure accelerates t-PA binding, alters binding affinity, and increases maximum binding to polymerized fibrin, effects that may result from ultrasound-induced changes in fibrin structure.
Collapse
|
19
|
Fujise K, Revelle BM, Stacy L, Madison EL, Yeh ET, Willerson JT, Beck PJ. A tissue plasminogen activator/P-selectin fusion protein is an effective thrombolytic agent. Circulation 1997; 95:715-22. [PMID: 9024162 DOI: 10.1161/01.cir.95.3.715] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND P-selectin is expressed on the surface of activated endothelial cells and platelets. We hypothesized that a tissue plasminogen activator (TPA)/P-selectin fusion protein would have not only thrombolytic activity but also might target TPA to the thrombi. In addition, it seemed possible that this chimeric protein would competitively inhibit the binding of native P-selectin on endothelial cells and platelets to leukocytes and thus further promote thrombolysis. METHODS AND RESULTS The full-length, plasminogen activator inhibitor-1-resistant form of TPA (TPAIR) together with two TPAIR/P-selectin fusion constructs (P280IR and P121IR) were expressed with the use of baculovirus vectors. After infection of Sf21 cells with the recombinant baculovirus, recombinant TPAIR and P-selectin/TPAIR fusion proteins were purified with the use of metal ion chromatography. The intact protease activity of TPAIR and the ligand binding capability of P-selectin were confirmed through indirect chromogenic and cell binding assays, respectively. These molecules were assessed both in vitro and in vivo for thrombolytic activity. In vitro clot lysis assays indicated equal efficacy of TPAIR, P280IR, and P121IR (P > .5). The in vivo efficacy was tested in a cyclic flow variation model with the use of the rat mesenteric artery. Compared with saline control treatment, reduction in cyclic flow variations was significant (P < .05) and similar (P > .5) among TPAIR, P280IR, and P121IR. No significant bleeding was noted among treated animals. CONCLUSIONS Chimeric proteins P280IR and P121IR have clot lysis activities that are similar to TPAIR both in vitro and in vivo. These chimeric proteins also bind to P-selectin ligand in vitro. Thus, these proteins may provide an efficient method of targeting TPA to the thrombotic region. Further experimental analysis with the use of larger animal coronary occlusion models should help determine the future value of these proteins as clinical therapeutic agents.
Collapse
Affiliation(s)
- K Fujise
- Department of Internal Medicine, University of Texas-Houston Health Science Center 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
Horrevoets AJ, Pannekoek H, Nesheim ME. A steady-state template model that describes the kinetics of fibrin-stimulated [Glu1]- and [Lys78]plasminogen activation by native tissue-type plasminogen activator and variants that lack either the finger or kringle-2 domain. J Biol Chem 1997; 272:2183-91. [PMID: 9036151 DOI: 10.1074/jbc.272.4.2183] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The kinetics of activation of both [Glu1]- and [Lys78]Plg(S741C-fluorescein by native (recombinant) tissue-type plasminogen activator and its deletion variants lacking either the finger or kringle-2 domain were measured by fluorescence within fully polymerized fibrin clots. The kinetics conform to the Michaelis-Menten equation at any fixed fibrin concentration so long as the plasminogen concentration is expressed as either the free or fibrin-bound, but not the total. The apparent kcat and Km values both vary systematically with the concentration of fibrin. Competition kinetics disclosed an active site-dependent interaction between t-Pa and [Glu1]Plg(S741C-fluorescein) in the presence, but not the absence, of fibrin. A steady-state template model having the rate equation v/[A]o = kcat(app).[Plg]/(Km(app) + [Plg]) was derived and used to interpret the data. The model indicates that catalytic efficiency is determined by the stability of the ternary activator-fibrin-plasminogen complex rather than the binding of the activator or plasminogen to fibrin. This implies that efforts to improve the enzymatic properties of t-PA might be more fruitfully directed at enhancing the stability of the ternary complex rather than fibrin binding.
Collapse
Affiliation(s)
- A J Horrevoets
- Department of Biochemistry, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | | | | |
Collapse
|
21
|
Runge MS, Harker LA, Bode C, Ruef J, Kelly AB, Marzec UM, Allen E, Caban R, Shaw SY, Haber E, Hanson SR. Enhanced thrombolytic and antithrombotic potency of a fibrin-targeted plasminogen activator in baboons. Circulation 1996; 94:1412-22. [PMID: 8823001 DOI: 10.1161/01.cir.94.6.1412] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Thrombolytic therapy reduces mortality in patients with acute myocardial infarction, but significant limitations exist with the use of currently available agents. In the present report, we describe the thrombolytic and antithrombotic potencies of a hybrid recombinant plasminogen activator consisting of an antifibrin antibody 59D8 (AFA) and low-molecular-weight single-chain urokinase-type plasminogen activator (scuPA). METHODS AND RESULTS A thrombolysis model in which thrombi are preformed in vivo in juvenile baboons was developed to compare the potencies of AFA-scuPA, recombinant tissue plasminogen activator (rTPA), and recombinant scuPA (rscuPA) in lysing nonocclusive 111In-labeled platelet-rich arterial-type thrombi and 125I-labeled fibrin-rich venous-type thrombi. Systemic infusion of 1.89 nmol/kg AFA-scuPA produced thrombolysis that was comparable to that obtained with much higher doses of TPA (14.2 nmol/kg) and rscuPA (28.5 nmol/kg). When steady-state plasma concentrations are normalized, AFA-scuPA lyses thrombi sixfold more rapidly than scuPA and TPA (P < .001) and reduces the rate of formation more than comparable doses of rscuPA (P < .0001). At equivalent thrombolytic doses, AFA-scuPA produced fewer antihemostatic effects than either rTPA or rscuPA. Template bleeding time measurements were shorter (3.5 +/- 0.12 minutes for AFA-scuPA versus 5.3 +/- 0.36 and 5.2 +/- 0.04 minutes for rTPA and rscuPA, respectively; P < .05), alpha 2-antiplasmin consumption was less (P < .05), and D-dimer generation was lower (P < .05). CONCLUSIONS We conclude that antibody targeting of scuPA to fibrin increases thrombolytic and antithrombotic potencies with less impairment of hemostasis compared with rTPA and rscuPA.
Collapse
Affiliation(s)
- M S Runge
- Cardiology Division, University of Texas Medical Branch, Galveston 77555-0553, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Smith JW, Tachias K, Madison EL. Protein loop grafting to construct a variant of tissue-type plasminogen activator that binds platelet integrin alpha IIb beta 3. J Biol Chem 1995; 270:30486-90. [PMID: 8530479 DOI: 10.1074/jbc.270.51.30486] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Protein-protein interactions can be guided by contacts between surface loops within proteins. We therefore investigated the hypothesis that novel protein-protein interactions could be created using a strategy of "loop grafting" in which the amino acid sequence of a biologically active, flexible loop on one protein is used to replace a surface loop present on an unrelated protein. To test this hypothesis we replaced a surface loop within an epidermal growth factor module with the complementarity-determining region of a monoclonal antibody. Specifically, the HCDR3 from Fab-9, an antibody selected to bind the beta 3-integrins with nanomolar affinity (Smith, J. W., Hu, D., Satterthwait, A., Pinz-Sweeney, S., and Barbas, C. F., III (1994) J. Biol. Chem. 269, 32788-32795), was grafted into the epidermal growth factor-like module of human tissue-type plasminogen activator (t-PA). The resulting variant of t-PA bound to the platelet integrin alpha IIb beta 3 with nanomolar affinity, retained full enzymatic activity, and was stimulated normally by the physiological co-factor fibrin. Binding of the novel variant of t-PA to integrin alpha IIb beta 3 was dependent on the presence of divalent cations and was inhibited by an RGD-containing peptide, demonstrating that, like the donor antibody, the novel t-PA binds specifically to the ligand-binding site of the integrin. These findings suggest that surface loops within protein modules can, at least in some cases, be interchangeable and that phage display can be combined with loop grafting to direct proteins, at high affinity, to selected targets. In principle, these targets could include not only other proteins but also peptides, nucleic acids, carbohydrates, lipids, or even uncharacterized markers of specific cell types, tissues, or viruses.
Collapse
Affiliation(s)
- J W Smith
- Department of Vascular Biology, Scripps Research Institute, La Jolla, California 92037, USA
| | | | | |
Collapse
|
23
|
Novokhatny V, Medved L, Lijnen HR, Ingham K. Tissue-type plasminogen activator (tPA) interacts with urokinase-type plasminogen activator (uPA) via tPA's lysine binding site. An explanation of the poor fibrin affinity of recombinant tPA/uPA chimeric molecules. J Biol Chem 1995; 270:8680-5. [PMID: 7721771 DOI: 10.1074/jbc.270.15.8680] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Differential scanning calorimetry was used to study the domain structure and intramolecular interactions of tPA/uPA chimeras. A high temperature transition centered near 90 degrees C was observed upon melting of the tPA/uPA chimera (amino acids 1-274 of tPA and 138-411 of uPA) and its variant lacking the finger and epidermal growth factor-like modules (residues 1-3 and 87-274 of tPA and 138-411 of uPA). Since neither of the two parent plasminogen activators display such a stable structure, one may suggest that a new stabilizing intramolecular interaction occurs in the chimeras. We found that occupation of the lysine binding site of tPA by a lysine or arginine side chain from the urokinase moiety is responsible for the high temperature transition as well as for the failure of the chimeras to exhibit the expected fibrin binding properties. All uPA species, single- and two-chain high molecular weight uPA (Pro-Uk and HMW-Uk) and two-chain low molecular weight uPA (LMW-Uk), interact intermolecularly with tPA and its kringle-containing derivatives. This intermolecular interaction was strongly inhibited by epsilon-aminocaproic acid indicating that the lysine binding site of tPA is involved. The binding of uPA with the fluorescein-labeled A-chain of tPA, registered by changes in fluorescence anisotropy, was estimated to have a Kd range of 1-7 microM. The interaction of tPA with uPA determined by solid-phase assays appeared to be tighter, with a Kd range of 50-300 nM. Two synthetic peptides, with and without carboxyl-terminal lysine, corresponding to urokinase residues 144-158 and 144-157, were approximately 100-fold more potent than epsilon-aminocaproic acid with respect to inhibition of the tPA-uPA interaction, indicating that the tPA binding site on urokinase is located within this sequence, close to the activation site Lys158-Ile159. The discovered intermolecular interaction may be related to the reported synergistic effect of simultaneous administration of these two plasminogen activators.
Collapse
Affiliation(s)
- V Novokhatny
- Holland Laboratory, American Red Cross, Rockville, Maryland 20855, USA
| | | | | | | |
Collapse
|
24
|
Kotkow KJ, Deitcher SR, Furie B, Furie BC. The second kringle domain of prothrombin promotes factor Va-mediated prothrombin activation by prothrombinase. J Biol Chem 1995; 270:4551-7. [PMID: 7876224 DOI: 10.1074/jbc.270.9.4551] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The incorporation of factor Xa into the prothrombinase complex, factor Xa-factor Va-phospholipid-Ca(II), results in an approximately 10(5)-fold higher rate of substrate activation than that of the enzyme alone. To examine the role of the prothrombin kringle domains in the interaction with prothrombinase we have employed site-directed mutagenesis to produce prothrombin species that lack either the first kringle domain, PT/delta K1, or the second kringle domain, PT/delta K2. Previously, we have shown that these proteins are fully carboxylated and that they bind to phospholipid vesicles. In this investigation we demonstrate that cleavage at Arg271-Thr272 and Arg320-Ile321 peptide bonds occurs upon activation with prothrombinase to yield normal thrombin from both PT/delta K1 and PT/delta K2. In the absence of factor Va, the Km(app) for the activation of PT/delta K1, PT/delta K2, or plasma-derived prothrombin by factor Xa-phospholipid-Ca(II) are equivalent. The Km(app) for the activation of PT/delta K2 by prothrombinase is approximately 4-5-fold higher than that obtained for plasma-derived prothrombin or PT/delta K1. These data demonstrate that the prothrombin kringle domains do not contribute significantly to the binding affinity of the substrate-enzyme interaction. In the absence of factor Va, equivalent kcat values were obtained for all of the prothrombin species when they were activated by factor Xa-Ca(II)-phospholipid. In contrast, a 7-fold lower kcat value was obtained for the activation of PT/delta K2 by prothrombinase as compared with that obtained for plasma prothrombin or PT/delta K1. Collectively, these data suggest that determinants within the second prothrombin kringle domain interact with factor Va to elicit a significant acceleration in the catalytic rate of substrate turnover. Indeed, in contrast to plasma-derived prothrombin, no direct binding of PT/delta K2 to factor Va to form the PT/delta K2-factor Va complex could be demonstrated by 90 degrees light scattering.
Collapse
Affiliation(s)
- K J Kotkow
- Division of Hematology-Oncology, New England Medical Center, Boston, Massachusetts
| | | | | | | |
Collapse
|
25
|
Yahara H, Matsumoto K, Maruyama H, Nagaoka T, Okada K, Ueshima S, Matsuo O. Recombinant variants of tissue-type plasminogen activator containing amino acid substitutions between positions 37 and 42. ACTA ACUST UNITED AC 1994. [DOI: 10.1016/0268-9499(94)90017-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
26
|
Stroughton J, Ouriel K, Shortell CK, Cho JS, Marder VJ. Plasminogen acceleration of urokinase thrombolysis. J Vasc Surg 1994; 19:298-303; discussion 303-5. [PMID: 8114191 DOI: 10.1016/s0741-5214(94)70105-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE A relative deficiency of plasminogen within the thrombus may be the rate limiting factor in clot lysis. METHODS To investigate this hypothesis, we used an in vitro perfusion system and expanded polytetrafluoroethylene graft segments filled with radiolabeled human thrombus. Three groups of five perfusions were compared: (1) urokinase infusion (333 IU/min) into clots laced with buffer, (2) urokinase infusion (333 IU/min) into clots laced with plasminogen (44 CU), and (3) control, D5W infusion into clots laced with buffer. Two end points were measured over time: the amount of lysed thrombus and the flow through the graft. RESULTS Urokinase infusion resulted in augmented flow through the graft when compared with control (p < 0.05). Lacing with plasminogen resulted in more rapid restoration of flow when compared with urokinase infusion alone (p < 0.05). Similarly, the rate of clot dissolution was significantly greater in plasminogen-laced thrombi (p < 0.05) when compared with the control and urokinase groups. Embolization of particles of thrombus was uniformly observed in the urokinase group, resulting in a temporary decrease in flow through the thrombosed graft. This event characteristically occurred after 60 minutes of infusion but was never seen in the urokinase/plasminogen treatment group. CONCLUSIONS These results suggest that plasminogen supplementation of urokinase thrombolysis may result in significant clinical benefits with respect to the rate of clot lysis and the uniformity of clot dissolution with a lower likelihood of secondary embolization.
Collapse
Affiliation(s)
- J Stroughton
- Department of Surgery, University of Rochester, NY 14642
| | | | | | | | | |
Collapse
|
27
|
Madison E. Probing structure-function relationships of tissue-type plasminogen activator by site-specific mutagenesis. ACTA ACUST UNITED AC 1994. [DOI: 10.1016/0268-9499(94)90720-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
28
|
Pharmacodynamics of modified tissue plasminogen activator LY210825 in the conscious dog as evaluated with immunocapture assays. ACTA ACUST UNITED AC 1993. [DOI: 10.1016/0268-9499(93)90135-i] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
29
|
Lubin I, Caban R, Runge M. The tissue plasminogen activator finger domain confers fibrin-dependent enhancement of catalytic activity to single-chain urokinase-type plasminogen activator. J Biol Chem 1993. [DOI: 10.1016/s0021-9258(18)53356-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
30
|
Yamashiro Y, Nakamura M, Huang GW, Kosugi T. Fibrin binding, fibrinolytic and fibrinogenolytic activity of plasminogen activator derived from the paranasal mucous membrane of humans. Laryngoscope 1993; 103:197-202. [PMID: 8426513 DOI: 10.1002/lary.5541030214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
It is known that large amounts of plasminogen activator (PA) are contained in tissue extracts of the human paranasal mucous membrane (PMM) with chronic sinusitis. The present study was undertaken to isolate and purify the PA in tissue extracts of PMM. Furthermore, the purified PA was identified as to whether it was of the tissue type or urokinase (UK) type, and some of its fibrinolytic characteristics were determined in comparison with those of urokinase. As starting material, extracts of acetone powder of PMM with chronic sinusitis were used, and Zn-imminodiacetate affinity chromatography, and ultrafiltration were carried out to separate and purify the PA from the PMM. The PA was purified to a 107-fold increase in specific activity. The molecular weight of the PA was estimated to be 65,000 to 70,000 d by gel filtration using Sephacryl S-200. The purified PA was stable in the range of pH 8.0 to 9.0. Using S-2288, a synthetic substrate, the Michaelis constant (Km) of the purified PA was estimated to be 0.11 mmol. The binding of the purified PA to fibrin was stronger than that of UK, while the fibrinogenolytic activity of the purified PA was not stronger than that of UK. Based on these results, the purified PA was identified as a tissue-type plasminogen activator (t-PA). From the kinetic data, it was identified as being of the two-chain variety. It is considered that, as a thrombolytic agent, t-PA derived from the PMM could be more useful than UK.
Collapse
Affiliation(s)
- Y Yamashiro
- Department of Physiology, School of Medicine, University of the Ryukyus, Okinawa, Japan
| | | | | | | |
Collapse
|
31
|
Madison EL, Sambrook JE. Probing structure-function relationships of tissue-type plasminogen activator by oligonucleotide-mediated site-specific mutagenesis. Methods Enzymol 1993; 223:249-71. [PMID: 8271957 DOI: 10.1016/0076-6879(93)23050-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- E L Madison
- Department of Vascular Biology, Scripps Research Institute, La Jolla, California 92037
| | | |
Collapse
|
32
|
Nguyen TH, Ward C. Stability characterization and formulation development of alteplase, a recombinant tissue plasminogen activator. PHARMACEUTICAL BIOTECHNOLOGY 1993; 5:91-134. [PMID: 8019701 DOI: 10.1007/978-1-4899-1236-7_3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- T H Nguyen
- Department of Pharmaceutical Research and Development, Genentech, Inc., South San Francisco, California 94080
| | | |
Collapse
|
33
|
Bassel-Duby R, Jiang N, Bittick T, Madison E, McGookey D, Orth K, Shohet R, Sambrook J, Gething M. Tyrosine 67 in the epidermal growth factor-like domain of tissue-type plasminogen activator is important for clearance by a specific hepatic receptor. J Biol Chem 1992. [DOI: 10.1016/s0021-9258(19)50142-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
34
|
Collen D, Lu HR, Lijnen HR, Nelles L, Stassen JM. Thrombolytic and pharmacokinetic properties of chimeric tissue-type and urokinase-type plasminogen activators. Circulation 1991; 84:1216-34. [PMID: 1909220 DOI: 10.1161/01.cir.84.3.1216] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Chimeric molecules comprising the A-chain of tissue-type plasminogen activator (t-PA) and the catalytic domain of urokinase-type plasminogen activator (u-PA) have intact enzymatic characteristics of u-PA, partial fibrin-binding properties of t-PA, and thrombolytic properties in animal models comparable with but not superior to those of single-chain u-PA (scu-PA). Deletion of the finger and growth factor domains (t-PA-delta FE/scu-PA-e) in such chimeras further reduces their affinity for fibrin. METHODS AND RESULTS A detailed investigation of the thrombolytic potency and the pharmacokinetics of t-PA and u-PA chimeras was performed in quantitative animal models for thrombolysis. In hamsters with pulmonary embolism, in rabbits with jugular vein thrombosis, and in baboons with femoral vein thrombosis, the thrombolytic potency (percent lysis per milligram of compound administered per kilogram of body weight) of t-PA-delta FE/scu-PA-e was significantly higher than that of recombinant scu-PA (rscu-PA, Saruplase) as shown by a maximal rate of 720 +/- 170% versus 45 +/- 5% lysis per milligram of compound per kilogram of body weight (mean +/- SEM, p less than 0.01) in hamsters, 210 +/- 18% versus 49 +/- 3% lysis per milligram of compound per kilogram of body weight (mean +/- SEM, p less than 0.01) in rabbits, and 310 +/- 73% versus 90 +/- 0.3% lysis per milligram of compound per kilogram of body weight (p less than 0.01) in baboons. However, the specific thrombolytic activity (percent lysis per microgram per milliliter steady-state plasma antigen level) of t-PA-delta FE/scu-PA-e was not significantly different from that of rscu-PA in hamsters (210 +/- 57% versus 160 +/- 27% lysis per microgram per milliliter antigen level) and was lower than that of rscu-PA in rabbits (37 +/- 4% versus 130 +/- 5% lysis per microgram per milliliter antigen level; p less than 0.01). In dogs with a combined femoral vein blood clot and a platelet-rich femoral arterial eversion graft thrombosis, 0.25 mg/kg body wt bolus injections of t-PA-delta FE/scu-PA-e produced significantly more venous clot lysis (90 +/- 5%, n = 10) than 0.25 mg/kg rscu-PA (26 +/- 3%, n = 10) (p less than 0.001) and, at the arterial side, more frequent (10 of 10 dogs versus three of 10 dogs) and more persistent (six of 10 dogs versus none of 10 dogs) recanalization (p = 0.002). After bolus injection in hamsters, rabbits, or baboons, t-PA-delta FE/scu-PA-e had a fourfold to sixfold longer initial half-life than rscu-PA and a slower plasma clearance of sixfold in hamsters, 10-fold in rabbits, and more than 10-fold in baboons. CONCLUSIONS These results indicate that t-PA-delta FE/scu-PA-e has a markedly enhanced thrombolytic potency toward venous and arterial thrombi caused by a delayed in vivo clearance with relatively maintained specific thrombolytic activity. These properties suggest that the chimera may be clinically useful for thrombolytic therapy by bolus administration in patients with thromboembolic disease.
Collapse
Affiliation(s)
- D Collen
- Center for Thrombosis and Vascular Research, University of Leuven, Belgium
| | | | | | | | | |
Collapse
|
35
|
Affiliation(s)
- D Collen
- Center for Thrombosis and Vascular Research, University of Leuven, Belgium
| | | | | |
Collapse
|
36
|
Interactions of plasminogen and tissue plasminogen activator (t-PA) with amphoterin. Enhancement of t-PA-catalyzed plasminogen activation by amphoterin. J Biol Chem 1991. [DOI: 10.1016/s0021-9258(18)55362-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
37
|
Novokhatny V, Ingham K, Medved L. Domain structure and domain-domain interactions of recombinant tissue plasminogen activator. J Biol Chem 1991. [DOI: 10.1016/s0021-9258(18)98794-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
38
|
Synthesis, purification and biological properties of a truncated mutant form of human tissue plasminogen activator produced in E. Coli. ACTA ACUST UNITED AC 1991. [DOI: 10.1016/0268-9499(91)90022-v] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
39
|
Larsen G, Timony G, Horgan P, Barone K, Henson K, Angus L, Stoudemire J. Protein engineering of novel plasminogen activators with increased thrombolytic potency in rabbits relative to activase. J Biol Chem 1991. [DOI: 10.1016/s0021-9258(18)92955-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
40
|
Preparation, in vivo properties and proposed clinical use of polyoxyethylene-modified tissue plasminogen activator and streptokinase. Adv Drug Deliv Rev 1991. [DOI: 10.1016/0169-409x(91)90038-e] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
41
|
Affiliation(s)
- H R Lijnen
- Center for Thrombosis and Vascular Research, K.U.L., Campus Gasthuisberg, Leuven, Belgium
| | | |
Collapse
|
42
|
Langer-Safer PR, Ahern TJ, Angus LB, Barone KM, Brenner MJ, Horgan PG, Morris GE, Stoudemire JB, Timony GA, Larsen GR. Replacement of finger and growth factor domains of tissue plasminogen activator with plasminogen kringle 1. Biochemical and pharmacological characterization of a novel chimera containing a high affinity fibrin-binding domain linked to a heterologous protein. J Biol Chem 1991. [DOI: 10.1016/s0021-9258(19)67853-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
43
|
Pohl G, Sterky C, Attersand A, Nyberg E, Löwenadler B, Hansson L. Tissue plasminogen activator mutants lacking the growth factor domain and the first kringle domain: I. ACTA ACUST UNITED AC 1991. [DOI: 10.1016/0268-9499(91)90074-e] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
44
|
Collen D, Bennett WF. Recombinant tissue-type plasminogen activator. BIOTECHNOLOGY (READING, MASS.) 1991; 19:197-223. [PMID: 1786472 DOI: 10.1016/b978-0-7506-9120-8.50015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
45
|
Nesheim M, Fredenburgh JC, Larsen GR. The dissociation constants and stoichiometries of the interactions of Lys-plasminogen and chloromethyl ketone derivatives of tissue plasminogen activator and the variant delta FEIX with intact fibrin. J Biol Chem 1990. [DOI: 10.1016/s0021-9258(18)45774-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
46
|
Morton P, Owensby D, Wun T, Billadello J, Schwartz A. Identification of determinants involved in binding of tissue-type plasminogen activator-plasminogen activator inhibitor type 1 complexes to HepG2 cells. J Biol Chem 1990. [DOI: 10.1016/s0021-9258(18)77272-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
47
|
Wilhelm O, Jaskunas S, Vlahos C, Bang N. Functional properties of the recombinant kringle-2 domain of tissue plasminogen activator produced in Escherichia coli. J Biol Chem 1990. [DOI: 10.1016/s0021-9258(18)77345-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
48
|
Trill J, Fong KL, Shebuski R, McDevitt P, Rosa M, Johanson K, Williams D, Boyle K, Sellers T, Reff M. Expression and characterisation of finger protease (FP); a mutant tissue-type plasminogen activator (t-PA) with improved pharmacokinetics. ACTA ACUST UNITED AC 1990. [DOI: 10.1016/s0268-9499(05)80044-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
49
|
Collen D, Lijnen HR, Bulens F, Vandamme AM, Tulinsky A, Nelles L. Biochemical and functional characterization of human tissue-type plasminogen activator variants with mutagenized kringle domains. J Biol Chem 1990. [DOI: 10.1016/s0021-9258(19)38329-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
50
|
Ahern TJ, Morris GE, Barone KM, Horgan PG, Timony GA, Angus LB, Henson KS, Stoudemire JB, Langer-Safer PR, Larsen GR. Site-directed mutagenesis in human tissue-plasminogen activator. Distinguishing sites in the amino-terminal region required for full fibrinolytic activity and rapid clearance from the circulation. J Biol Chem 1990. [DOI: 10.1016/s0021-9258(19)39394-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|