1
|
Boro N, Alexandrino Fernandes P, Mukherjee AK. Computational analysis to comprehend the structure-function properties of fibrinolytic enzymes from Bacillus spp for their efficient integration into industrial applications. Heliyon 2024; 10:e33895. [PMID: 39055840 PMCID: PMC11269858 DOI: 10.1016/j.heliyon.2024.e33895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Background The fibrinolytic enzymes from Bacillus sp. are proposed as therapeutics in preventing thrombosis. Computational-based analyses of these enzymes' amino acid composition, basic physiological properties, presence of functional domain and motifs, and secondary and tertiary structure analyses can lead to developing a specific enzyme with improved catalytic activity and other properties that may increase their therapeutic potential. Methods The nucleotide sequences of fibrinolytic enzymes produced by the genus Bacillus and its corresponding protein sequences were retrieved from the NCBI database and aligned using the PRALINE programme. The varied physiochemical parameters and structural and functional analysis of the enzyme sequences were carried out with the ExPASy-ProtParam tool, MEME server, SOPMA, PDBsum tool, CYS-REC tool, SWISS-MODEL, SAVES servers, TMHMM program, GlobPlot, and peptide cutter software. The assessed in-silico data were compared with the published experimental results for validation. Results The alignment of sixty fibrinolytic serine protease enzymes (molecular mass 12-86 kDa) sequences showed 49 enzymes possess a conserved domain with a catalytic triad of Asp196, His242, and Ser569. The predicted instability and aliphatic indexes were 1.94-37.77, and 68.9-93.41, respectively, indicating high thermostability. The random coil means value suggested the predominance of this secondary structure in these proteases. A set of 50 amino acid residues representing motif 3 signifies the Peptidase S8/S53 domain that was invariably observed in 56 sequences. Additionally, 28 sequences have transmembrane helices, with two having the most disordered areas, and they pose 25 enzyme cleavage sites. A comparative analysis of the experimental work with the results of in-silico study put forward the characteristics of the enzyme sequences JF739176.1 and MF677779.1 to be considered when creating a potential mutant enzyme as these sequences are stable at high pH with thermostability and to exhibit αβ-fibrinogenase activity in both experimental and in-silico studies.
Collapse
Affiliation(s)
- Nitisha Boro
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028, Assam, India
| | - Pedro Alexandrino Fernandes
- LAQV@REQUIMTE, Departamento de Química e Bioquímica, Faculdade De Ciências, Universidade do Porto, Rua Do Campo Alegre S/N, 4169-007, Porto, Portugal
| | - Ashis K. Mukherjee
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028, Assam, India
- Microbial Biotechnology and Protein Research Laboratory, Division of Life Sciences, Institute of Advanced Studies in Science and Technology, Vigyan Path, Garchuk, Paschim Boragaon, Guwahati, 781035, Assam, India
| |
Collapse
|
2
|
Rehan R, Virk S, Wong CCY, Passam F, Layland J, Keech A, Yong A, White HD, Fearon W, Ng M. Intracoronary thrombolysis in ST-elevation myocardial infarction: a systematic review and meta-analysis. Heart 2024; 110:988-996. [PMID: 38925881 PMCID: PMC11287581 DOI: 10.1136/heartjnl-2024-324078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Despite restoration of epicardial blood flow in acute ST-elevation myocardial infarction (STEMI), inadequate microcirculatory perfusion is common and portends a poor prognosis. Intracoronary (IC) thrombolytic therapy can reduce microvascular thrombotic burden; however, contemporary studies have produced conflicting outcomes. OBJECTIVES This meta-analysis aims to evaluate the efficacy and safety of adjunctive IC thrombolytic therapy at the time of primary percutaneous coronary intervention (PCI) among patients with STEMI. METHODS Comprehensive literature search of six electronic databases identified relevant randomised controlled trials. The primary outcome was major adverse cardiac events (MACE). The pooled risk ratio (RR) and weighted mean difference (WMD) with a 95% CI were calculated. RESULTS 12 studies with 1915 patients were included. IC thrombolysis was associated with a significantly lower incidence of MACE (RR=0.65, 95% CI 0.51 to 0.82, I2=0%, p<0.0004) and improved left ventricular ejection fraction (WMD=1.87; 95% CI 1.07 to 2.67; I2=25%; p<0.0001). Subgroup analysis demonstrated a significant reduction in MACE for trials using non-fibrin (RR=0.39, 95% CI 0.20 to 0.78, I2=0%, p=0.007) and moderately fibrin-specific thrombolytic agents (RR=0.62, 95% CI 0.47 to 0.83, I2=0%, p=0.001). No significant reduction was observed in studies using highly fibrin-specific thrombolytic agents (RR=1.10, 95% CI 0.62 to 1.96, I2=0%, p=0.75). Furthermore, there were no significant differences in mortality (RR=0.91; 95% CI 0.48 to 1.71; I2=0%; p=0.77) or bleeding events (major bleeding, RR=1.24; 95% CI 0.47 to 3.28; I2=0%; p=0.67; minor bleeding, RR=1.47; 95% CI 0.90 to 2.40; I2=0%; p=0.12). CONCLUSION Adjunctive IC thrombolysis at the time of primary PCI in patients with STEMI improves clinical and myocardial perfusion parameters without an increased rate of bleeding. Further research is needed to optimise the selection of thrombolytic agents and treatment protocols.
Collapse
Affiliation(s)
- Rajan Rehan
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Concord Hospital, Concord, New South Wales, Australia
| | - Sohaib Virk
- Systematic Reviews, CORE Group, Sydney, New South Wales, Australia
| | - Christopher C Y Wong
- Cardiology, Concord Repatriation General Hospital, Concord, New South Wales, Australia
- Stanford Hospital, Stanford, California, USA
| | - Freda Passam
- Department of Hematology, University of Sydney, Sydney, New South Wales, Australia
| | | | - Anthony Keech
- NHMRC Clinical Trials Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Andy Yong
- Cardiology, Concord Repatriation General Hospital, Concord, New South Wales, Australia
| | - Harvey D White
- Cardiology Department, Green Lane Cardiovascular Service and Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand
| | | | - Martin Ng
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Department of Cardiology, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
Raghunathan S, Rayes J, Sen Gupta A. Platelet-inspired nanomedicine in hemostasis thrombosis and thromboinflammation. J Thromb Haemost 2022; 20:1535-1549. [PMID: 35435322 PMCID: PMC9323419 DOI: 10.1111/jth.15734] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 12/01/2022]
Abstract
Platelets are anucleate cell-fragments derived predominantly from megakaryocytes in the bone marrow and released in the blood circulation, with a normal count of 150 000-40 000 per μl and a lifespan of approximately 10 days in humans. A primary role of platelets is to aid in vascular injury site-specific clot formation to stanch bleeding, termed hemostasis. Platelets render hemostasis by a complex concert of mechanisms involving platelet adhesion, activation and aggregation, coagulation amplification, and clot retraction. Additionally, platelet secretome can influence coagulation kinetics and clot morphology. Therefore, platelet defects and dysfunctions result in bleeding complications. Current treatment for such complications involve prophylactic or emergency transfusion of platelets. However, platelet transfusion logistics constantly suffer from limited donor availability, challenges in portability and storage, high bacterial contamination risks, and very short shelf life (~5 days). To address these issues, an exciting area of research is focusing on the development of microparticle- and nanoparticle-based platelet surrogate technologies that can mimic various hemostatic mechanisms of platelets. On the other hand, aberrant occurrence of the platelet mechanisms lead to the pathological manifestation of thrombosis and thromboinflammation. The treatments for this are focused on inhibiting the mechanisms or resolving the formed clots. Here, platelet-inspired technologies can provide unique platforms for disease-targeted drug delivery to achieve high therapeutic efficacy while avoiding systemic side-effects. This review will provide brief mechanistic insight into the role of platelets in hemostasis, thrombosis and thromboinflammation, and present the current state-of-art in the design of platelet-inspired nanomedicine for applications in these areas.
Collapse
Affiliation(s)
- Shruti Raghunathan
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| | - Julie Rayes
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Anirban Sen Gupta
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
4
|
Padrick MM, Brown W, Lyden PD. Intravenous Thrombolysis. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00053-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
5
|
Mican J, Toul M, Bednar D, Damborsky J. Structural Biology and Protein Engineering of Thrombolytics. Comput Struct Biotechnol J 2019; 17:917-938. [PMID: 31360331 PMCID: PMC6637190 DOI: 10.1016/j.csbj.2019.06.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/25/2019] [Accepted: 06/27/2019] [Indexed: 12/22/2022] Open
Abstract
Myocardial infarction and ischemic stroke are the most frequent causes of death or disability worldwide. Due to their ability to dissolve blood clots, the thrombolytics are frequently used for their treatment. Improving the effectiveness of thrombolytics for clinical uses is of great interest. The knowledge of the multiple roles of the endogenous thrombolytics and the fibrinolytic system grows continuously. The effects of thrombolytics on the alteration of the nervous system and the regulation of the cell migration offer promising novel uses for treating neurodegenerative disorders or targeting cancer metastasis. However, secondary activities of thrombolytics may lead to life-threatening side-effects such as intracranial bleeding and neurotoxicity. Here we provide a structural biology perspective on various thrombolytic enzymes and their key properties: (i) effectiveness of clot lysis, (ii) affinity and specificity towards fibrin, (iii) biological half-life, (iv) mechanisms of activation/inhibition, and (v) risks of side effects. This information needs to be carefully considered while establishing protein engineering strategies aiming at the development of novel thrombolytics. Current trends and perspectives are discussed, including the screening for novel enzymes and small molecules, the enhancement of fibrin specificity by protein engineering, the suppression of interactions with native receptors, liposomal encapsulation and targeted release, the application of adjuvants, and the development of improved production systems.
Collapse
Key Words
- EGF, Epidermal growth factor domain
- F, Fibrin binding finger domain
- Fibrinolysis
- K, Kringle domain
- LRP1, Low-density lipoprotein receptor-related protein 1
- MR, Mannose receptor
- NMDAR, N-methyl-D-aspartate receptor
- P, Proteolytic domain
- PAI-1, Inhibitor of tissue plasminogen activator
- Plg, Plasminogen
- Plm, Plasmin
- RAP, Receptor antagonist protein
- SAK, Staphylokinase
- SK, Streptokinase
- Staphylokinase
- Streptokinase
- Thrombolysis
- Tissue plasminogen activator
- Urokinase
- t-PA, Tissue plasminogen activator
Collapse
Affiliation(s)
- Jan Mican
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Martin Toul
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - David Bednar
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Jiri Damborsky
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| |
Collapse
|
6
|
Kirtane AR, Sadhukha T, Kim H, Khanna V, Koniar B, Panyam J. Fibrinolytic Enzyme Cotherapy Improves Tumor Perfusion and Therapeutic Efficacy of Anticancer Nanomedicine. Cancer Res 2017; 77:1465-1475. [PMID: 28108516 DOI: 10.1158/0008-5472.can-16-1646] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 12/17/2016] [Accepted: 12/21/2016] [Indexed: 12/14/2022]
Abstract
Elevated interstitial fluid pressure and solid stress within tumors contribute to poor intratumoral distribution of nanomedicine. In this study, we hypothesized that the presence of fibrin in tumor extracellular matrix contributes to hindered intratumoral distribution of nanocarriers and that this can be overcome through the use of a fibrinolytic enzyme such as tissue plasminogen activator (tPA). Analysis of fibrin expression in human tumor biopsies showed significant fibrin staining in nearly all tumor types evaluated. However, staining was heterogeneous across and within tumor types. We determined the effect of fibrin on the diffusion, intratumoral distribution, and therapeutic efficacy of nanocarriers. Diffusivity of nanocarriers in fibrin matrices was limited and could be improved significantly by coincubation with tPA. In vivo, coadministration of tPA improved the anticancer efficacy of nanoparticle-encapsulated paclitaxel in subcutaneous syngeneic mouse melanoma and orthotopic xenograft lung cancer models. Furthermore, treatment with tPA led to decompression of blood vessels and improved tumor perfusion. Cotreatment with tPA resulted in greater intratumoral penetration of a model nanocarrier (Doxil), leading to enhanced availability of the drug in the tumor core. Fibrinolytics such as tPA are already approved for other indications. Fibrinolytic cotherapy is therefore a rapidly translatable strategy for improving therapeutic effectiveness of anticancer nanomedicine. Cancer Res; 77(6); 1465-75. ©2017 AACR.
Collapse
Affiliation(s)
- Ameya R Kirtane
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota
| | - Tanmoy Sadhukha
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota
| | - Hyunjoon Kim
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota
| | - Vidhi Khanna
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota
| | - Brenda Koniar
- Research Animal Resources, University of Minnesota, Minneapolis, Minnesota
| | - Jayanth Panyam
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota. .,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
7
|
Choi JH, Lee HJ, Kim S. Purification and antithrombotic activity of wulfase, a fibrinolytic enzyme from the fruit bodies of the edible and medicinal mushroom Sparassis crispa Wulf. ex. Fr. APPL BIOCHEM MICRO+ 2016. [DOI: 10.1134/s000368381606003x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Babu V, Subathra Devi C. Exploring the in vitro thrombolytic potential of streptokinase-producing β-hemolytic Streptococci isolated from bovine milk. J GEN APPL MICROBIOL 2016; 61:139-46. [PMID: 26377134 DOI: 10.2323/jgam.61.139] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The aim of this study was to isolate and characterize streptokinase-producing β-hemolytic Streptococcus sp. from bovine milk. A total of 50 milk samples were collected randomly from different breeds of cow and goat (Vellore, Tamil Nadu, India). The samples were characterized and screened for streptokinase-producing isolates using microbial and biochemical analysis. About 97 colonies were isolated from milk samples showing hemolytic patterns of α (19.6%), β (24.7%) and γ (55.6 %). Out of 20β-hemolytic isolates, only 6 colonies (VB2, VB3, VB8, VB14, VB16, and VB17) were identified as β-hemolytic Streptococci as potent producers of streptokinase. VB2 and VB14 showed the greatest streptokinase activities of 265 U mL(-1) and 225 U mL(-1), respectively. Based on biochemical and molecular characterization, the potent isolates VB2 and VB14 were identified and confirmed as S. equinus and S. agalactiae, respectively. The identified strains were named Streptococcus equinus VIT_VB2 (GenBank accession no. JX406835) and Streptococcus agalactiae VITVS5 (GenBank accession No. KF186620) The strains isolated from bovine milk provide a variance in the fibrinolytic activity on blood clots. The current study has demonstrated that the isolation of streptokinase producers from bovine milk, and the production of streptokinase from novel strain, enhanced the fibrinolytic activity. This study is the first to report that Streptococcus equinus produces streptokinase.
Collapse
|
9
|
Hidaka K, Gohda K, Teno N, Wanaka K, Tsuda Y. Active site-directed plasmin inhibitors: Extension on the P2 residue. Bioorg Med Chem 2016; 24:545-53. [DOI: 10.1016/j.bmc.2015.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/04/2015] [Accepted: 12/07/2015] [Indexed: 10/22/2022]
|
10
|
Intravenous Thrombolysis. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00051-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
|
11
|
Mirzapoiazova T, Mambetsariev N, Lennon FE, Mambetsariev B, Berlind JE, Salgia R, Singleton PA. HABP2 is a Novel Regulator of Hyaluronan-Mediated Human Lung Cancer Progression. Front Oncol 2015; 5:164. [PMID: 26258071 PMCID: PMC4508840 DOI: 10.3389/fonc.2015.00164] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/06/2015] [Indexed: 01/08/2023] Open
Abstract
Background Lung cancer is a devastating disease with limited treatment options. Many lung cancers have changes in their microenvironment including upregulation of the extracellular matrix glycosaminoglycan, hyaluronan (HA), which we have previously demonstrated can regulate the activity of the extracellular serine protease, hyaluronan binding protein 2 (HABP2). This study examined the functional role of HABP2 on HA-mediated human lung cancer dynamics. Methods Immunohistochemical analysis was performed on lung cancer patient samples using anti-HABP2 antibody. Stable control, shRNA, and HABP2 overexpressing human lung adenocarcinoma cells were evaluated using immunoblot analysis, migration, extravasation, and urokinase plasminogen activator (uPA) activation assays with or without high-molecular weight HA or low-molecular weight HA (LMW-HA). In human lung cancer xenograft models, primary tumor growth rates and lung metastasis were analyzed using consecutive tumor volume measurements and nestin immunoreactivity in nude mouse lungs. Results We provide evidence that HABP2 is an important regulator of lung cancer progression. HABP2 expression was increased in several subtypes of patient non-small cell lung cancer samples. Further, HABP2 overexpression increased LMW-HA-induced uPA activation, migration, and extravasation in human lung adenocarcinoma cells. In vivo, overexpression of HABP2 in human lung adenocarcinoma cells increased primary tumor growth rates in nude mice by ~2-fold and lung metastasis by ~10-fold compared to vector control cells (n = 5/condition). Conclusion Our data suggest a possible direct effect of HABP2 on uPA activation and lung cancer progression. Our observations suggest that exploration of HABP2 in non-small cell lung carcinoma merits further study both as a diagnostic and therapeutic option.
Collapse
Affiliation(s)
- Tamara Mirzapoiazova
- Section of Pulmonary and Critical Care, Department of Medicine, Pritzker School of Medicine, The University of Chicago , Chicago, IL , USA
| | - Nurbek Mambetsariev
- Section of Pulmonary and Critical Care, Department of Medicine, Pritzker School of Medicine, The University of Chicago , Chicago, IL , USA
| | - Frances E Lennon
- Section of Pulmonary and Critical Care, Department of Medicine, Pritzker School of Medicine, The University of Chicago , Chicago, IL , USA ; Section of Hematology/Oncology, Department of Medicine, Pritzker School of Medicine, The University of Chicago , Chicago, IL , USA
| | - Bolot Mambetsariev
- Section of Pulmonary and Critical Care, Department of Medicine, Pritzker School of Medicine, The University of Chicago , Chicago, IL , USA
| | - Joshua E Berlind
- Section of Pulmonary and Critical Care, Department of Medicine, Pritzker School of Medicine, The University of Chicago , Chicago, IL , USA
| | - Ravi Salgia
- Section of Hematology/Oncology, Department of Medicine, Pritzker School of Medicine, The University of Chicago , Chicago, IL , USA
| | - Patrick A Singleton
- Section of Pulmonary and Critical Care, Department of Medicine, Pritzker School of Medicine, The University of Chicago , Chicago, IL , USA ; Department of Anesthesia and Critical Care, Pritzker School of Medicine, The University of Chicago , Chicago, IL , USA
| |
Collapse
|
12
|
Abstract
There has been a significant amount of research done on liposomes and nanoparticles as drug carriers for protein drugs. Proteins and enzymes have been used both as targeting moieties and for their therapeutic potential. High specificity and rapid reaction rates make proteins and enzymes excellent candidates for therapeutic treatment, but some limitations exist. Many of these limitations can be addressed by a well studied nanotechnology based delivery system. Such a system can provide a medium for delivery, stabilization of the drugs, and enable site specific accumulation of drugs. Nanomedicines such as these have great potential to revolutionize the pharmaceutical industry and improve healthcare worldwide.
Collapse
Affiliation(s)
- John N Barry
- Department of Bioengineering, Clemson University, 301 Rhodes Hall, Clemson, SC 29634, United States,
| | - Alexey A Vertegel
- Department of Bioengineering, Clemson University, 301 Rhodes Hall, Clemson, SC 29634, United States,
| |
Collapse
|
13
|
Bondada L, Rondla R, Pradere U, Liu P, Li C, Bobeck D, McBrayer T, Tharnish P, Courcambeck J, Halfon P, Whitaker T, Amblard F, Coats SJ, Schinazi RF. Azetidines and spiro azetidines as novel P2 units in hepatitis C virus NS3 protease inhibitors. Bioorg Med Chem Lett 2013; 23:6325-30. [PMID: 24135727 DOI: 10.1016/j.bmcl.2013.09.068] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 09/19/2013] [Accepted: 09/23/2013] [Indexed: 12/14/2022]
Abstract
Herein, we report the synthesis and structure-activity relationship studies of new analogs of boceprevir 1 and telaprevir 2. Introduction of azetidine and spiroazetidines as a P2 substituent that replaced the pyrrolidine moiety of 1 and 2 led to the discovery of a potent hepatitis C protease inhibitor 37c (EC50=0.8 μM).
Collapse
Affiliation(s)
- Lavanya Bondada
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Veterans Affairs Medical Center, 1670 Haygood Drive, NE, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Bilheiro RP, Braga AD, Filho ML, Carvalho-Tavares J, Agero U, Carvalho MDG, Sanchez EF, Salas CE, Lopes MTP. The thrombolytic action of a proteolytic fraction (P1G10) from Carica candamarcensis. Thromb Res 2013; 131:e175-82. [PMID: 23473638 DOI: 10.1016/j.thromres.2013.01.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 01/18/2013] [Accepted: 01/22/2013] [Indexed: 01/03/2023]
Abstract
A group of cysteine-proteolytic enzymes from C. candamarcensis latex, designated as P1G10 displays pharmacological properties in animal models following various types of lesions. This enzyme fraction expresses in vitro fibrinolytic effect without need for plasminogen activation. Based on this evidence, we assessed by intravital microscopy the effect of P1G10 on recanalization of microvessels after thrombus induction in the ear of hairless mice. Video playback of intravital microscopic images allowed measurement of blood flow velocity (mm/s) during the experimental procedure. Groups treated with 5 or 7.5mg/Kg P1G10 showed thrombolysis between 7-15min, without vessel obstruction. Ex vivo experiments demonstrated that platelet activation by ADP is impaired in a dose dependent manner following treatment with P1G10. The P1G10 action on plasma coagulation also showed that prothrombin time (PT), thrombin time (TT) and activated partial thromboplastin time (aPTT, μg/uL) are increased in a dose dependent manner. In addition, P1G10 displayed fibrinogenolytic and fibrinolytic activities, both in a dose dependent manner. Each of these effects was suppressed by inhibition of the proteolytic activity of the fraction. The antithrombotic action of P1G10 can be explained by proteolytic cleavage of fibrinogen and fibrin, both key factors during formation of a stable thrombus. These results combined with prior evidence suggest that P1G10 has potential as thrombolytic agent.
Collapse
Affiliation(s)
- Rogério P Bilheiro
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
|
16
|
Thrombolytic, anticoagulant and antiplatelet activities of codiase, a bi-functional fibrinolytic enzyme from Codium fragile. Biochimie 2013; 95:1266-77. [PMID: 23402909 DOI: 10.1016/j.biochi.2013.01.023] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 01/31/2013] [Indexed: 11/23/2022]
Abstract
Thrombosis is a leading cause of morbidity and mortality throughout the world. Thrombolytic agents are important for both the prevention and treatment of thrombosis. In this study, codiase, a new bi-functional fibrinolytic serine protease having thrombolytic, anticoagulant, and antiplatelet activities was purified from marine green alga, Codium fragile. The molecular weight of the enzyme was estimated to be 48.9 kDa by SDS-PAGE, and mass spectrometry. Fibrin zymography analysis showed an active band with similar molecular weight. The N-terminal sequence was found to be APKASTDQTLPL, which is different from that of other known fibrinolytic enzymes. Codiase displayed maximum activity at 30 °C and pH 6.0, and the activity was inhibited by Zn(2+) and Fe(2+). Moreover, the enzyme activity was strongly inhibited by serine protease inhibitor such as PMSF. Codiase exhibited high specificity for the substrate S-2288, and the Km and Vmax values for this substrate were found to be 0.24 mM and 79 U/ml respectively. Fibrin plate assays revealed that it was able to hydrolyze fibrin clot either directly or by activation of plasminogen. Codiase effectively hydrolyzed fibrin and fibrinogen, preferentially degrading α- and Aα chains, followed by γ-γ, and γ-chains. However, it provoked slower degradation of Bβ and β-chains. The structural change of fibrin clot and fibrinogen by codiase was also detected by FTIR-ATR spectroscopy analysis. In vitro and in vivo studies revealed that codiase reduces thrombosis in concentration-dependent manner. Codiase was found to prolong activated partial thromboplastin time (APTT), and prothrombin time (PT). PFA-100 studies showed that codiase prolonged the closure time (CT) of citrated whole human blood. These favorable antithrombotic profiles together with its anticoagulant and platelet disaggregation properties, and lack of toxicity to mice and NIH-3T3 cells, make it a potential agent for thrombolytic therapy.
Collapse
|
17
|
Vachharajani NN, Raymond RH, Shyu WC, Stouffer BC, Boulton DW. The effects of age and gender on the pharmacokinetics and pharmacodynamics in healthy subjects of the plasminogen activator, lanoteplase. Br J Clin Pharmacol 2011; 72:775-86. [PMID: 21545481 PMCID: PMC3243012 DOI: 10.1111/j.1365-2125.2011.04003.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 04/27/2011] [Indexed: 11/29/2022] Open
Abstract
AIMS To investigate the influence of age and gender on the intravenous pharmacokinetics and pharmacodynamics of the plasminogen activator, lanoteplase. METHODS Forty healthy subjects (10 each of young males, elderly males, young females and elderly females) received a single bolus 10 kU kg(-1) intravenous dose of lanoteplase. Plasma from blood serially collected for 24 h post-dose was analyzed for lanoteplase (antigen), fibrinogen, plasminogen and α2-antiplasmin concentrations, plasma plasminogen activation activity (PPAA) and rapid plasminogen activator inhibitor (PAI-1). RESULTS Lanoteplase mean total systemic clearance (CL(t)) values ranged from 1.9 to 2.8 l h(-1) and mean steady-state volume of distribution (V(ss)) values ranged from 12.3 to 15.6 l. Age-by-gender interactions were observed for lanoteplase CL(t) (P= 0.04), but no differences were observed for V(ss) or elimination half-life. Elderly females had a 27% lower mean CL(t) than young females (95% CI for the difference 0.17, 1.27 l h(-1)) and 32% lower CL(t) than elderly males (95% CI for the difference 0.15, 1.65 l h(-1)). PPAA AUC/dose values did not show an age-by-gender interaction. Haemostasis parameters indicated only a slight degree of systemic plasminogen activation. CONCLUSIONS Elderly females had a lower mean lanoteplase CL(t) than elderly males and young females. However, no difference was observed between young and elderly females for the AUC/dose of PPAA. In addition, there were no age-related or gender-related differences observed in the other pharmacodynamic parameters measured.
Collapse
Affiliation(s)
- Nimish N Vachharajani
- Bristol-Myers Squibb Research and Development, Princeton, New Jersey 08543-4000, USA
| | | | | | | | | |
Collapse
|
18
|
Abstract
Studies in animal models have shown that plasminogen activators bound to erythrocytes (RBC-PA) have an extended lifetime in the circulation and are safer than free PAs. RBC-PAs incorporate into nascent thrombi, which are focally lysed from within, an attractive thromboprophylactic option. In static systems, RBC-PAs cleave surrounding fibrin fibers, forming pores larger than the cells themselves, and move around the pore edges, enlarging them until eventual clot dissolution. We hypothesized that under flow in blood vessels, RBC-PAs form functional patent channels before clot dissolution. Here we used perfusion chambers to study clot lysis by RBC-PAs under static versus arterial and venous flow conditions. We found that flow decelerates bulk clot lysis but quickly generates patent channels filled with passing RBCs, via pore enlargement and merging in the direction of flow. Formation of such channels by RBC-PAs may help rescue ischemic tissue before bulk dissolution of potentially occlusive clots.
Collapse
|
19
|
Brown W, Al-Khoury L, Tafreshi G, Lyden PD. Intravenous Thrombolysis. Stroke 2011. [DOI: 10.1016/b978-1-4160-5478-8.10049-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
20
|
Sustained thromboprophylaxis mediated by an RBC-targeted pro-urokinase zymogen activated at the site of clot formation. Blood 2010; 115:5241-8. [PMID: 20410503 DOI: 10.1182/blood-2010-01-261610] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Plasminogen activators (PAs) are used to treat life-threatening thrombosis, but not for thromboprophylaxis because of rapid clearance, risk of bleeding, and central nervous system (CNS) toxicity. We describe a novel strategy that may help to overcome these limitations by targeting a thrombin-activated PA pro-drug to circulating red blood cells (RBCs). We fused a single chain antibody (scFv Ter-119) that binds to mouse glycophorin A (GPA) with a variant human single-chain low molecular weight urokinase construct that can be activated selectively by thrombin (scFv/uPA-T). scFv/uPA-T bound specifically to mouse RBCs without altering their biocompatibility and retained its zymogenic properties until converted by thrombin into an active 2-chain molecule. As a result, RBC-bound scFv/uPA-T caused thrombin-induced fibrinolysis. One hour and 48 hours after intravenous (IV) injection in mice, approximately 70% and approximately 35% of scFv/uPA-T was retained in the blood, respectively, and approximately 95% of the circulating scFv/uPA-T remained bound to RBCs. A single IV injection of scFv/uPA-T provided effective prophylaxis against arterial and venous thrombosis for up to 24 hours. Thus, prophylactic delivery of RBC-targeted PA pro-drugs activated selectively at the site of clot formation represents a new approach to prevent thrombosis in clinical settings where the risk of clotting is high.
Collapse
|
21
|
Zaitsev S, Zaitzev S, Spitzer D, Murciano JC, Ding BS, Tliba S, Kowalska MA, Bdeir K, Kuo A, Stepanova V, Atkinson JP, Poncz M, Cines DB, Muzykantov VR. Targeting of a mutant plasminogen activator to circulating red blood cells for prophylactic fibrinolysis. J Pharmacol Exp Ther 2009; 332:1022-31. [PMID: 19952305 DOI: 10.1124/jpet.109.159194] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chemical coupling to carrier red blood cells (RBCs) converts tissue type plasminogen activator (tPA) from a problematic therapeutic into a safe agent for thromboprophylaxis. The goal of this study was to develop a more clinically relevant recombinant biotherapeutic by fusing a mutant tPA with a single-chain antibody fragment (scFv) with specificity for glycophorin A (GPA) on mouse RBCs. The fusion construct (anti-GPA scFv/PA) bound specifically to mouse but not human RBCs and activated plasminogen; this led to rapid and stable attachment of up to 30,000 copies of anti-GPA scFv/PA per mouse RBC that were thereby endowed with high fibrinolytic activity. Binding of anti-GPA scFv/PA neither caused RBC aggregation, hemolysis, uptake in capillary-rich lungs or in the reticuloendothelial system nor otherwise altered the circulation of RBCs. Over 40% of labeled anti-GPA scFv/PA injected in mice bound to RBC, which markedly prolonged its intravascular circulation and fibrinolytic activity compared with its nontargeted PA counterpart, anti-GPA scFv/PA, but not its nontargeted PA analog, prevented thrombotic occlusion in FeCl(3) models of vascular injury. These results provide proof-of-principle for the development of a recombinant PA variant that binds to circulating RBC and provides thromboprophylaxis by use of a clinically relevant approach.
Collapse
Affiliation(s)
- Sergei Zaitsev
- IFEM, University of Pennsylvania School of Medicine, One John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA 19104-6068, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Yurko Y, Maximov V, Andreozzi E, Thompson GL, Vertegel AA. Design of biomedical nanodevices for dissolution of blood clots. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2009. [DOI: 10.1016/j.msec.2009.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
23
|
Ghazali M, Hayward GL. Acoustic determination of performance and equivalence of plasminogen activators. Anal Bioanal Chem 2008; 392:897-902. [PMID: 18759103 DOI: 10.1007/s00216-008-2343-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Revised: 07/17/2008] [Accepted: 08/07/2008] [Indexed: 11/30/2022]
Abstract
A reliable method for the measurement of different plasminogen activators is of great interest for both manufacturing and clinical medicine. A one-step assay based on a thickness shear mode acoustic sensor has been developed for this purpose. Two separate mixtures of substrates (fibrinogen and plasminogen) and enzymes (thrombin and the plasminogen activator) were mixed, and placed on the acoustic sensor surface. During the assay, the resonant frequency of a quartz crystal oscillating in the thickness shear mode was measured and used to find a characteristic clot dissolution time, from the sample addition to the time at the maximum dissolution rate. Calibrations of the acoustic assay were done for tissue-type plasminogen activator (t-PA) as well as for the other plasminogen activators: urokinase (u-PA); streptokinase (SK) and staphylokinase (SAK). All gave relative standard deviations of about 12%. Since the same method was used for all of the activators, their activities were compared, resolving the differences between their unit definitions. Linear relationships were found between urokinase and streptokinase which activate plasminogen directly and between t-PA and staphylokinase which require fibrin as a cofactor. The relationship between the groups was found to curve, indicating the difference between the two mechanisms. The acoustic method, therefore, may be used as a rapid and cost-effective reference method for the standardization and comparison of different plasminogen activators.
Collapse
Affiliation(s)
- Mirnader Ghazali
- Faculty of Engineering, University of Isfahan, Isfahan, 81746-73441, Iran.
| | | |
Collapse
|
24
|
Ghazali M, Hayward GL. One-step thickness shear mode acoustic assay for plasminogen activators. Analyst 2008; 133:910-3. [PMID: 18575644 DOI: 10.1039/b718187f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A new procedure is presented for the measurement of plasminogen activators using a thickness shear mode sensor and a modified version of the fibrin plate assay at the micro-scale. Separate, well-mixed solutions of the substrates fibrinogen and plasminogen, and enzymes thrombin and the plasminogen activator sample were mixed together and placed on the sensor surface. The temperature and evaporation were controlled during the assay. The clot dissolution time correlated well with the quantity of the plasminogen activator in the sample. The average relative standard deviation was 12.5%.
Collapse
Affiliation(s)
- Mirnader Ghazali
- Faculty of Engineering, University of Isfahan, Isfahan, 81746-73441, Iran.
| | | |
Collapse
|
25
|
Agero U, Arantes RME, Lacerda-Queiroz N, Mesquita ON, Magalhães A, Sanchez EF, Carvalho-Tavares J. Effect of mutalysin II on vascular recanalization after thrombosis induction in the ear of the hairless mice model. Toxicon 2007; 50:698-706. [PMID: 17681580 DOI: 10.1016/j.toxicon.2007.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 06/01/2007] [Accepted: 06/04/2007] [Indexed: 10/23/2022]
Abstract
Mutalysin II (mut-II) is an alpha-fibrinogenase isolated from Lachesis muta muta (bushmaster) snake venom. The enzyme lyses fibrin clots in vitro, and this activity does not depend on plasminogen activation. The aim of this study was to assess by intravital microscopy the effect of Mutalysin II on the recanalization of microvessels after thrombus induction in the ears of hairless mice. Photochemical thrombus formation was induced after i.v. injection of 5% fluorescein isothiocyanate labelled dextran (FITC-dextran) followed by mercury light exposure of individual microvessels of the ear of five anesthetized animals. Video playback analysis of intravital microscopy images of the ear microcirculation permitted us to measure blood flow velocity (microm/s) under control conditions (before thrombus formation) in the ear microvessels. Thirty minutes after thrombus formation (blood flow velocity stopped completely), each animal (n=5) was infused with Mutalysin II (2.0 mg/kg, i.v.). All animals treated with Mutalysin II showed evident thrombolysis after approximately 12 min, followed by recanalization. A separate group of mice (n=5) which received urokinase type-plasminogen activator (u-PA, 250 U/mouse, i.v.) showed blood flow restoration within the same interval (12 min). These in vivo data suggest that Mutalysin II has the potential to be an effective thrombolytic agent.
Collapse
Affiliation(s)
- Ubirajara Agero
- Departamento de Física, Instituto de Ciência Exatas Universidade Federal de Minas Gerais, Brazil
| | | | | | | | | | | | | |
Collapse
|
26
|
Hitosugi M, Omura K, Yufu T, Kido M, Niwa M, Nagai T, Tokudome S. Changes in blood viscosity with the recombinant tissue plasminogen activator alteplase. Thromb Res 2007; 120:447-50. [PMID: 17064755 DOI: 10.1016/j.thromres.2006.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2005] [Revised: 09/16/2006] [Accepted: 09/18/2006] [Indexed: 11/20/2022]
Abstract
We measured whole blood viscosity to investigate the time course of the fibrinolytic activity of the recombinant tissue plasminogen activator alteplase. Changes in blood viscosity over time were determined using an oscillation-type viscometer at a shear rate of 400 to 500 per second. Blood viscosity initially increased with alteplase as in untreated blood, but then decreased, reflecting the fibrinolytic activity of generated plasmin. Blood viscosity subsequently stabilized at a level below the initial value owing to the dissolution of both fibrin and fibrinogen by alteplase. To our knowledge, the present study is the first to examine the time course of changes in BV during fibrin formation and degradation. The results indicated that the fibrinolyic agent alteplase might provide the additional benefit of increasing blood flow by lowering blood viscosity.
Collapse
Affiliation(s)
- Masahito Hitosugi
- Department of Legal Medicine, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Mibu, Shimotsuga, Tochigi 321-0293, Japan.
| | | | | | | | | | | | | |
Collapse
|
27
|
Hagemeyer CE, Tomic I, Weirich U, Graeber J, Nordt T, Runge MS, Bode C, Peter K. Construction and characterization of a recombinant plasminogen activator composed of an anti-fibrin single-chain antibody and low-molecular-weight urokinase. J Thromb Haemost 2004; 2:797-803. [PMID: 15099287 DOI: 10.1111/j.1538-7836.2004.00697.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Targeting of plasminogen activators to the fibrin component of a thrombus by antibodies directed against human fibrin can enhance their thrombolytic potency and clot specificity. OBJECTIVES To overcome the disadvantages of chemical conjugation, we investigated whether the recombinant fusion of a single-chain antibody and a plasminogen activator results in an active bifunctional molecule that might be useful as a therapeutic agent. METHODS The cDNA of low-molecular-weight single-chain urokinase-type plasminogen activator, comprising amino acids Leu144-Leu411 (scuPA(LMW)), was cloned from human endothelial cells and fused to a single-chain antibody specific for the 7 N-terminal amino acids (beta(15-22)) in the beta-chain of human fibrin (scFv(59D8)). The fusion protein was purified using affinity chromatography with the beta(15-22)-peptide of human fibrin. RESULTS Purified scFv(59D8)-scuPA(LMW) migrated as a 60-kDa band, which is consistent with a molecule composed of one scFv(59D8) and one scuPA(LMW) moiety. Both functions of the fusion molecule, fibrin-specific binding and plasminogen activation, were fully preserved. In human plasma clots, thrombolysis by scFv(59D8)-scuPA(LMW) is significantly faster and more potent compared with the clinically used urokinase. CONCLUSIONS ScFv(59D8)-scuPA(LMW) constitutes a new recombinant chimeric plasminogen activator with a significantly enhanced thrombolytic potency and relative fibrin selectivity, that can be produced with modern methods at low cost, large quantities and reproducible activity in Escherichia coli.
Collapse
Affiliation(s)
- C E Hagemeyer
- Department of Cardiology, University of Freiburg, Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Al-Khoury L, Lyden PD. Intravenous Thrombolysis. Stroke 2004. [DOI: 10.1016/b0-44-306600-0/50056-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
29
|
Murciano JC, Medinilla S, Eslin D, Atochina E, Cines DB, Muzykantov VR. Prophylactic fibrinolysis through selective dissolution of nascent clots by tPA-carrying erythrocytes. Nat Biotechnol 2003; 21:891-6. [PMID: 12845330 DOI: 10.1038/nbt846] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2003] [Accepted: 05/23/2003] [Indexed: 11/09/2022]
Abstract
A fibrinolytic agent consisting of a tissue-type plasminogen activator (tPA) coupled to the surface of red blood cells (RBCs) can dissolve nascent clots from within the clot, in a Trojan horse-like strategy, while having minimal effects on preexisting hemostatic clots or extravascular tissue. After intravenous injection, the fibrinolytic activity of RBC-tPA persisted in the bloodstream at least tenfold longer than did that of free tPA. In a model of venous thrombosis induced by intravenously injected fibrin microemboli aggregating in pulmonary vasculature, soluble tPA lysed pulmonary clots lodged before but not after tPA injection, whereas the converse was true for RBC-tPA. Free tPA failed to lyse occlusive carotid thrombosis whether injected before or after vascular trauma, whereas RBC-tPA circulating before, but not injected after, thrombus formation restored blood flow. This RBC-based drug delivery strategy alters the fibrinolytic profile of tPA, permitting prophylactic fibrinolysis.
Collapse
Affiliation(s)
- Juan-Carlos Murciano
- Institute for Environmental Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | |
Collapse
|
30
|
|
31
|
Moons L, Vanlinthout I, Roelants I, Moreadith R, Collen D, Rapold HJ. Toxicology studies with recombinant staphylokinase and with SY 161-P5, a polyethylene glycol-derivatized cysteine-substitution mutant. Toxicol Pathol 2001; 29:285-91. [PMID: 11442014 DOI: 10.1080/019262301316905237] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
SY 161-P5, a polyethylene glycol derivatized (PEGylated) mutant of the recombinant Staphylokinase (rSak) variant SakSTAR, exhibiting reduced antigenicity is in clinical development for treatment of acute myocardial infarction as a single bolus injection. A series of safety studies were performed in vivo as a routine toxicology program with SY 161-P5 (PEG-rSakSTAR) and with the recombinant Staphylokinase variant Sak42D (rSak42D). For both compounds, intravenous single bolus injections of up to 100-fold therapeutic equivalent, as well as repeated injections during 7 to 28 days revealed no significant pathological findings in mice, rats or hamsters. However, New Zealand white rabbits developed clinically silent, multifocal myocarditis following single or repeat doses of SY 161-P5 or of Sak42D. These findings were dose-independent and reversible. A similar species-specific cardiotoxic effect has previously been described for other proteolytic proteins, including the approved drugs Streptokinase and Acetylated Plasminogen Streptokinase Complex (APSAC). The large experience with these drugs, as well as the clinical data accumulated both with PEGylated and non-PEGylated rSak variants to date, do not indicate cardiotoxic hazards associated with the use of these drugs in humans.
Collapse
Affiliation(s)
- L Moons
- Center for Transgene Technology and Gene Therapy, Flanders Interuniversity Institute for Biotechnology, KU Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
32
|
Collen D. Ham-Wasserman lecture: role of the plasminogen system in fibrin-homeostasis and tissue remodeling. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2001; 2001:1-9. [PMID: 11722975 DOI: 10.1182/asheducation-2001.1.1] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Plasminogen can be converted to plasmin either via the tissue-type plasminogen activator (t-PA) or via the urokinase-type plasminogen activator (u-PA)/u-PA receptor (u-PAR) pathway. A dual role for these pathways is now well established: 1) t-PA is involved in fibrin homeostasis and 2) u-PA is primarily involved in cell migration and tissue remodeling. t-PA mediated activation is used for thrombolytic therapy of acute myocardial infarction and some other thromboembolic diseases. The u-PA mediated pathway, in concert with the matrix metalloproteinase (MMP) system, plays a pleiotropic role in arterial neointima formation, atherosclerosis, angiogenesis, tumor growth metastasis, and infarction. However, therapeutic interventions in the u-PA/MMP system remain to be further defined.
Collapse
Affiliation(s)
- D Collen
- Afdeling Moleculaire & Vasculaire Biologie, K.U. Leuven, O & N Gasthuisberg, Herestr. 49, B-3000 Leuven, Belgium
| |
Collapse
|
33
|
Tiffany BR, Barrali R. Advances in the pharmacology of acute coronary syndrome. Platelet inhibition. Emerg Med Clin North Am 2000; 18:723-43, vi. [PMID: 11130935 DOI: 10.1016/s0733-8627(05)70155-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The development of potent inhibitors of platelet aggregation has led to significant decreases in morbidity and mortality rates among patients undergoing percutaneous coronary intervention. Clinical trials have demonstrated that agents that block glycoprotein IIb/IIIa receptor-mediated platelet aggregation have an outcome benefit when used acutely in patients with chest pain and ST depression or elevated cardiac enzymes, leading to the integration of these agents into emergency medicine clinical practice. This article provides an overview of the pathophysiology of acute coronary syndrome and the pharmacology of platelet inhibition and reviews the evidence from the clinical trials pertaining to the use of these agents in the emergency department.
Collapse
Affiliation(s)
- B R Tiffany
- Department of Emergency Medicine, Maricopa Medical Center, Phoenix, Arizona, USA
| | | |
Collapse
|
34
|
Llevadot J, Guigliano RP. Pharmacology and clinical trial results of lanoteplase in acute myocardial infarction. Expert Opin Investig Drugs 2000; 9:2689-94. [PMID: 11060830 DOI: 10.1517/13543784.9.11.2689] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
New bolus fibrinolytic agents derived from the recombinant human tissue plasminogen activator (t-PA) have emerged as a new means of dissolution of the occlusive thrombosis associated with acute myocardial infarction. Lanoteplase is a fibrinolytic drug derived from t-PA by deleting its fibronectin finger-like and epidermal growth factor domains and mutating Asn(117) to Gln(117). Lanoteplase has a reduced plasma clearance and a prolonged half-life such that it can be administered as a single bolus. In the InTIME I trial, patency (TIMI grade 2 or 3 flow) with the 120 KU/kg dose was higher compared with front-loaded t-PA. The InTIME II trial demonstrated that lanoteplase was as effective as alteplase with regard to mortality. However, the rate of intracranial haemorrhage was significantly higher in lanoteplase-treated patients and further development of this compound has been halted.
Collapse
Affiliation(s)
- J Llevadot
- Cardiovascular Division, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
35
|
Abstract
Site directed mutagenesis (350 variants) of recombinant staphylokinase (SakSTAR), a potent fibrin-selective thrombolytic agent, was undertaken in order to reduce its antigenicity while maintaining its potency. Variants with K35A, (ie, Lys[K] in position 35 substituted with Ala[A]), E65D or E65Q, K74R or K74Q, E80A+D82A, K130T, and K135R displayed increased enzymatic activity or reduced binding of human staphylokinase-specific antibodies. Additive mutagenesis identified 8 variants with intact thrombolytic potencies, which absorbed down to less than a third of SakSTAR-specific antibodies. Intra-arterial administration in 61 patients with peripheral arterial occlusion caused no significant allergic reactions. Median neutralizing antibody titers (with 15 to 85 percentiles), expressed as microgram (μg) compound neutralized per milliliter plasma, were 4.4 (0.3 to 49) for the variants, compared with 12 (4 to 100) in 70 patients given wild-type SakSTAR (P = .002 by Mann-Whitney rank sum test). Overt neutralizing antibody induction (more than 5 μg compound neutralized per milliliter plasma) was observed in 57 of 70 patients (81%) given wild-type SakSTAR, but only in 28 of 60 patients (47%) treated with variants (P < .0001 by Fisher exact test). On the basis of this study, the variant SakSTAR (K35A, E65Q, K74R, D82A, S84A, T90A, E99D, T101S, E108A, K109A, K130T, K135R) (code SY155) has been selected for further clinical development.
Collapse
|
36
|
Recombinant staphylokinase variants with reduced antigenicity due to elimination of B-lymphocyte epitopes. Blood 2000. [DOI: 10.1182/blood.v96.4.1425] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Site directed mutagenesis (350 variants) of recombinant staphylokinase (SakSTAR), a potent fibrin-selective thrombolytic agent, was undertaken in order to reduce its antigenicity while maintaining its potency. Variants with K35A, (ie, Lys[K] in position 35 substituted with Ala[A]), E65D or E65Q, K74R or K74Q, E80A+D82A, K130T, and K135R displayed increased enzymatic activity or reduced binding of human staphylokinase-specific antibodies. Additive mutagenesis identified 8 variants with intact thrombolytic potencies, which absorbed down to less than a third of SakSTAR-specific antibodies. Intra-arterial administration in 61 patients with peripheral arterial occlusion caused no significant allergic reactions. Median neutralizing antibody titers (with 15 to 85 percentiles), expressed as microgram (μg) compound neutralized per milliliter plasma, were 4.4 (0.3 to 49) for the variants, compared with 12 (4 to 100) in 70 patients given wild-type SakSTAR (P = .002 by Mann-Whitney rank sum test). Overt neutralizing antibody induction (more than 5 μg compound neutralized per milliliter plasma) was observed in 57 of 70 patients (81%) given wild-type SakSTAR, but only in 28 of 60 patients (47%) treated with variants (P < .0001 by Fisher exact test). On the basis of this study, the variant SakSTAR (K35A, E65Q, K74R, D82A, S84A, T90A, E99D, T101S, E108A, K109A, K130T, K135R) (code SY155) has been selected for further clinical development.
Collapse
|
37
|
Affiliation(s)
- H R Lijnen
- Center for Molecular and Vascular Biology, University of Leuven, Belgium.
| | | |
Collapse
|
38
|
|
39
|
Pharmacokinetic and thrombolytic properties of cysteine-linked polyethylene glycol derivatives of staphylokinase. Blood 2000. [DOI: 10.1182/blood.v95.3.936.003k20_936_942] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recombinant staphylokinase (SakSTAR) variants obtained by site-directed substitution with cysteine, in the core (lysine 96 [Lys96], Lys102, Lys109, and/or Lys135) or the NH2-terminal region that is released during activation of SakSTAR (serine 2 [Ser2] and/or Ser3), were derivatized with thiol-specific (ortho-pyridyl-disulfide or maleimide) polyethylene glycol (PEG) molecules with molecular weights of 5000 (P5), 10 000 (P10), or 20 000 (P20). The specific activities and thrombolytic potencies in human plasma were unaltered for most variants derivatized with PEG (PEGylates), but maleimide PEG derivatives had a better temperature stability profile. In hamsters, SakSTAR was cleared at 2.2 mL/min; variants with 1 P5 molecule were cleared 2-to 5-fold; variants with 2 P5 or 1 P10 molecules were cleared 10-to 30-fold; and variants with 1 P20 molecule were cleared 35-fold slower. A bolus injection induced dose-related lysis of a plasma clot, fibrin labeled with 125 iodine (125I-fibrin plasma clot), and injected into the jugular vein. A 50% clot lysis at 90 minutes required 110 μg/kg SakSTAR; 50 to 110 μg/kg of core-substitution derivatives with 1 P5; 25 μg/kg for NH2-terminal derivatives with 1 P5; 5 to 25 μg/kg with derivatives with 2 P5 or 1 P10; and 7 μg/kg with P20 derivatives. Core substitution with 1 or 2 P5 molecules did not significantly reduce the immunogenicity of SakSTAR in rabbits. Derivatization of staphylokinase with a single PEG molecule allows controllable reduction of the clearance while maintaining thrombolytic potency at a reduced dose. This indicates that mono-PEGylated staphylokinase variants may be used for single intravenous bolus injection.
Collapse
|
40
|
Maksimenko AV, Tischenko EG. New thrombolytic strategy: bolus administration of tPA and urokinase-fibrinogen conjugate. J Thromb Thrombolysis 1999; 7:307-12. [PMID: 10373713 DOI: 10.1023/a:1008939428688] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Increased efficacy of thrombolytic therapy requires a comprehensive search for new and novel therapeutic strategies. Many new modified forms of plasminogen activators have been obtained by means of chemical and biological synthesis. However, clinical findings demonstrate that the reperfusion level achieved during thrombolysis remains the same for various thrombolytic agents, irrespective of an extensive search for an "ideal" thrombolytic. Thrombolytic therapy may be complicated by treatment delays, cumbersome schemes of preparation and administration, and hemorrhagic and rethrombotic events. These limitations may be overcome, at least in part, by applying combined thrombolysis with plasminogen activators exhibiting complementary actions and different pharmacokinetic profiles. The combined action of native thrombolytics allows the use of lower doses and simplified schemes of administration, yielding encouraging results in experimental models. Long-acting forms of plasminogen activators are being developed and tested in combination with tissue-type plasminogen activator as a trigger of thrombolysis. The combination of short- and long-acting plasminogen activators appears promising and potentially eligible for bolus administration to patients. On the basis of our own experimental results and data in the literature, we suggest a new thrombolytic strategy connected with the single injection of a combination of complementary and pharmacokinetically different plasminogen activators.
Collapse
Affiliation(s)
- A V Maksimenko
- Laboratory of Biochemical Engineering, Department of Biotechnology, Institute of Experimental Cardiology, Russian Cardiology Research Center, Moscow, Russia
| | | |
Collapse
|
41
|
Maksimenko AV, Tischenko EG, Golubykh VL. Thrombolytic effect after sequential double-bolus administration of tissue plasminogen activator and urokinase-fibrinogen conjugate into dogs with venous thrombosis. Ann N Y Acad Sci 1998; 864:409-17. [PMID: 9928118 DOI: 10.1111/j.1749-6632.1998.tb10349.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- A V Maksimenko
- Institute of Experimental Cardiology, Russian Cardiology Research Center, Moscow, Russia
| | | | | |
Collapse
|
42
|
Maksimenko AV. New strategy of thrombolysis. Conjunctive effect of plasminogen activators with different pharmacokinetic profile. Ann N Y Acad Sci 1998; 864:96-105. [PMID: 9928084 DOI: 10.1111/j.1749-6632.1998.tb10292.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Combined actions of native and prolonged thrombolytics allow the use of lower doses and simplified schemes of administration, thus yielding significant results in experimental therapy regarding the efficacy and safety of thrombolysis. Development of prolonged forms of plasminogen activators and testing their effect in combination with the thrombolysis trigger are well founded and of current interest. Thrombolytic compositions on the basis of short- and long-term-acting plasminogen activators appear to be promising and potentially eligible for bolus administration.
Collapse
Affiliation(s)
- A V Maksimenko
- Institute of Experimental Cardiology, Russian Cardiology Research Center, Moscow, Russia
| |
Collapse
|
43
|
|
44
|
Kamper EF, Kopeikina L, Mantas A, Stefanadis C, Toutouzas P, Stavridis J. Tetranectin levels in patients with acute myocardial infarction and their alterations during thrombolytic treatment. Ann Clin Biochem 1998; 35 ( Pt 3):400-7. [PMID: 9635106 DOI: 10.1177/000456329803500309] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Tetranectin (TN), a new regulator of fibrinolysis, was studied in the plasma of 60 patients with acute myocardial infarction (AMI) and 30 healthy subjects (HS), in relation to D-dimer (DD) and alpha 2-plasmin inhibitor (alpha 2-PI), to investigate its possible involvement in the pathophysiology of AMI. Thirty patients underwent thrombolytic treatment with fibrin-specific plasminogen activator (rt-PA) (group A); the other 30 patients, according to the exclusion criteria, were conventionally treated (group B). Twenty of the thrombolysized patients established early recanalization (subgroup A1), while 10 failed to respond to thrombolytic treatment (subgroup A2). Median (interquartile range), baseline plasma TN levels were lower in AMI patients compared to HS [8.27 (2.75) mg/L versus 12.1 (0.55) mg/L, P < 10(-6)]. In subgroup A1, TN increased at the end of rt-PA infusion and returned to the baseline levels 12 h later. A positive association between DD and TN release (3 h level minus baseline level) was found (rs = 0.48, P = 0.03) in subgroup A1. No significant alterations of TN levels were observed during therapy in subgroup A2 and group B. TN, DD and alpha 2-PI concentrations in group B remained relatively constant during the study period. This study provides evidence of a significant decrease of TN levels in AMI patients compared to healthy subjects and of a remarkable difference in the evolution of TN levels during thrombolytic treatment with rt-PA between recanalized and non-recanalized AMI patients. Thus, an involvement of TN in the formation and dissolution of fibrin clot in AMI patients is worthy of further investigation.
Collapse
Affiliation(s)
- E F Kamper
- Department of Experimental Physiology, Medical School, University of Athens, Greece
| | | | | | | | | | | |
Collapse
|
45
|
Thrombolysis strategy: Joint action of plasminogen activators. Thrombolytic compositions (A review). Pharm Chem J 1998. [DOI: 10.1007/bf02464204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
46
|
Maksimenko AV, Tishchenko EG, Dobrovol'skii AB. In vitro plasminogen activation by joint action of the tissue plasminogen activator and a urokinase—Ffbrinogen conjugate. Pharm Chem J 1998. [DOI: 10.1007/bf02464202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
47
|
Sitges M, Bosch X, Betriu A. [Optimization of thrombolytic treatment in acute myocardial infarct: the role of new fibrinoselective drugs and their combination with new antithrombotics]. Rev Esp Cardiol 1998; 51:178-91. [PMID: 9577163 DOI: 10.1016/s0300-8932(98)74731-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although reperfusion therapy is well recognized as the mainstay of treatment of acute myocardial infarction, mortality of myocardial infarction is still high, thrombolytic treatment remains underutilized and, usually, applied too late. Additionally, most of the patients do not experience optimal reperfusion because of the suboptimal flow rate in the infarct-related artery, abnormal microvascular flow, and reocclusion of the infarct-related artery. Strategies to enhance the results of reperfusion therapy include, expanding the population of potential candidates, earlier treatment, and newer methods to improve infarct-related artery flow rates. In this sense, new thrombolytic agents, and combination therapies with or without addition of more potent and specific new antithrombotic agents are being extensively investigated. Also, it is important to promote studies of ancillary treatments to reduce reperfusion injury, which may be one cause of decreased microvascular flow. Although aspirin and heparin have been the conventionally used agents for inhibiting thrombin and platelet function, newer agents such as hirudin or hirulog and inhibitors of the platelet glycoprotein IIb-IIIa receptors are becoming available, and their clinical application will increase in the future.
Collapse
Affiliation(s)
- M Sitges
- Institut de Malalties Cardiovasculars, Hospital Clínic, Universidad de Barcelona
| | | | | |
Collapse
|
48
|
Affiliation(s)
- D Collen
- Center for Molecular and Vascular Biology, University of Leuven, Belgium.
| |
Collapse
|
49
|
Abstract
Unstable angina and myocardial infarction (MI) continue to present a major challenge in clinical management. These acute ischemic coronary syndromes (AICS) are a spectrum of clinical presentations of the same pathophysiologic mechanism: thrombus formation superimposed on atherosclerotic plaque disruption or erosion. Current approaches to the management of AICS, which include both interventional and pharmacologic therapy, have been introduced to clinical practice during the past 20 years, and most of them have demonstrated efficacy in clinical studies. A common inadequacy of current therapies, however, is the lack of significant inhibition of platelet aggregation--the crucial event in the formation of coronary thrombi and the pathogenesis of AICS. The final common pathway to platelet aggregation is the activation of the platelet glycoprotein (GP) IIb-IIIa receptor, which allows the cross-linking of adjacent platelets by the adhesive plasma proteins fibrinogen and von Willebrand's factor. The emergence of the GP IIb-IIIa receptor as a potential treatment target has led to the development of several inhibitors of its function. The inhibitors most advanced in clinical development are the chimeric monoclonal antibody abciximab (ReoPro) and the cyclic peptide eptifibatide (INTEGRILIN). In phase III clinical trials, both abciximab and eptifibatide have been shown to reduce the incidence of cardiac events in patients at risk for abrupt vessel closure after coronary angioplasty. Inhibition of the GP IIb-IIIa receptor is the most promising novel approach to the treatment of unstable angina and MI, and it may soon be an indispensable component of the management of patients with AICS.
Collapse
Affiliation(s)
- H D White
- Cardiology Department, Green Lane Hospital, Auckland, New Zealand
| |
Collapse
|
50
|
de Bono D. Alteplase. Expert Opin Investig Drugs 1997; 6:1095-8. [PMID: 15989666 DOI: 10.1517/13543784.6.8.1095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Alteplase (recombinant human tissue type plasminogen activator, rt-PA) was identified as a naturally occurring plasminogen activator in 1975, cloned from a human melanoma cell line in 1981, introduced into clinical trials in 1982, and received a product licence for the treatment of acute myocardial infarction in 1986. The present review will concentrate on assessing its current status as a clinically effective thrombolytic agent, but will touch briefly on its biochemical, physiological and pharmacological properties. Since alteplase is an approved pharmaceutical name, the term tPA will be used to refer to the naturally occurring substance in its physiological context.
Collapse
Affiliation(s)
- D de Bono
- University of Leicester, Faculty of Medicine, Clinical Sciences Wing, Glenfield General Hospital, Leicester, LE3 9QP, UK
| |
Collapse
|