1
|
Grinstein J, Torii R, Salerno C, Blanco PJ, Garcia-Garcia H, Bourantas CV. Flow Dynamics During the HeartWare HVAD to HeartMate 3 Exchange: A Computational Study Assessing Differential Graft Lengths and Surgical Techniques. JTCVS Tech 2022; 14:125-126. [PMID: 35967208 PMCID: PMC9366874 DOI: 10.1016/j.xjtc.2022.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Jonathan Grinstein
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, Ill
| | - Ryo Torii
- Department of Engineering, University College of London, London, United Kingdom
| | - Christopher Salerno
- Department of Surgery, Section of Cardiothoracic Surgery, University of Chicago, Chicago, Ill
| | - Pablo J Blanco
- Department of Mathematical and Computational Methods, National Laboratory for Scientific Computing, Petrópolis, Brazil
| | - Hector Garcia-Garcia
- Department of Medicine, Section of Cardiology, MedStar Cardiovascular Research Network, Washington, DC
| | - Christos V Bourantas
- Department of Medicine, Section of Cardiology, Barts Heart Centre, London, United Kingdom
| |
Collapse
|
2
|
Mangin PH, Gardiner EE, Nesbitt WS, Kerrigan SW, Korin N, Lam WA, Panteleev MA. In vitro flow based systems to study platelet function and thrombus formation: Recommendations for standardization: Communication from the SSC on Biorheology of the ISTH. J Thromb Haemost 2020; 18:748-752. [PMID: 32112535 DOI: 10.1111/jth.14717] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/12/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022]
Abstract
Experimental videomicroscopic in vitro assays of thrombus formation based on blood perfusion are instrumental in a wide range of basic studies in thrombosis, screening for hereditary or acquired plateletrelated pathologies, and assessing the effectiveness of novel anti-platelet therapies. Here, we discuss application of the broadly used "in vitro thrombosis model": a frequently used assay to study the formation of 3D aggregates under flow, which involves perfusing anticoagulated whole blood over fibrillar collagen in a flow geometry of rectangular cross-section, such as glass microcapillaries or parallel-plate flow chambers. Major advantaged of this assay are simplicity and ability to reproduce the four main stages of platelet thrombus formation, i.e. platelet tethering, adhesion, activation and aggregation under a wide range of hemodynamic conditions. On the other hand, these devices represent, at best, simple reductive models of thrombosis. We also describe how blood flow assays can be used to study various aspects of platelet function on adhesive proteins and discuss the relevance of such flow models. Finally, we propose recommendations for standardization related to the use of this assay that cover collagen source, coating methods, micropatterning, sample composition, anticoagulation, choice of flow device, hemodynamic conditions, quantification challenges, variability, pre-analytical conditions and other issues.
Collapse
Affiliation(s)
- Pierre H Mangin
- INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, Université de Strasbourg, Strasbourg, France
| | - Elizabeth E Gardiner
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Warwick S Nesbitt
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
- School of Engineering, RMIT University, Melbourne, Victoria, Australia
| | - Steven W Kerrigan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Netanel Korin
- Faculty of Biomedical Engineering, Techion-Israel Institute of Technology, Haifa, Israel
| | - Wilbur A Lam
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta/Emory University School of Medicine, Atlanta, GA, USA
| | - Mikhail A Panteleev
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
3
|
Abstract
Fully bioresorbable scaffolds (BRS) were designed to overcome the limitations of metallic drug-eluting stents, which permanently cage the vessel wall, thereby preventing normal coronary vasomotion, preclude bypass grafting and can provoke long-term foreign-body responses. Although multiple scaffolds have been or are in development, the Absorb Bioresorbable Vascular Scaffold (BVS; Abbott Vascular) was the first FDA-approved device and was widely expected to fulfil the dream of interventional cardiologists of a transient scaffold that would disappear 'when the job was done' and would not hamper further treatment options. Although early, small studies and even large, randomized trials showed beneficial outcomes up to 1 year of follow-up, longer-term results have been disappointing, with increased rates of device thrombosis and target-lesion revascularization. The Absorb BVS device was withdrawn from the market because of low demand. In this Review, we summarize the preclinical and clinical data available for BRS to understand how the vascular biological reactions to these devices differ from biological reactions to metallic drug-eluting stents and how these responses translate into clinical outcomes. We also discuss next-generation BRS and outline modifications that are needed to improve the long-term outcomes with these devices so that they eventually become a viable option for patients with symptomatic obstructive coronary artery disease.
Collapse
|
4
|
Thromboprophylaxis strategies for children with single-ventricle circulations (superior or total cavo-pulmonary connections) after stent implantation. Cardiol Young 2019; 29:877-884. [PMID: 31208476 DOI: 10.1017/s1047951119000969] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To define optimal thromboprophylaxis strategy after stent implantation in superior or total cavopulmonary connections. BACKGROUND Stent thrombosis is a rare complication of intravascular stenting, with a perceived higher risk in single-ventricle patients. METHODS All patients who underwent stent implantation within superior or total cavopulmonary connections (caval vein, innominate vein, Fontan, or branch pulmonary arteries) were included. Cohort was divided into aspirin therapy alone versus advanced anticoagulation, including warfarin, enoxaparin, heparin, or clopidogrel. Primary endpoint was in-stent or downstream thrombus, and secondary endpoints included bleeding complications. RESULTS A total of 58 patients with single-ventricle circulation underwent 72 stent implantations. Of them 14 stents (19%) were implanted post-superior cavopulmonary connection and 58 (81%) post-total cavopulmonary connection. Indications for stenting included vessel/conduit stenosis (67%), external compression (18%), and thrombotic occlusion (15%). Advanced anticoagulation was prescribed for 32 (44%) patients and aspirin for 40 (56%) patients. Median follow up was 1.1 (25th-75th percentile, 0.5-2.6) years. Echocardiograms were available in 71 patients (99%), and advanced imaging in 44 patients (61%). Thrombosis was present in two patients on advanced anticoagulation (6.3%) and none noted in patients on aspirin (p = 0.187). Both patients with in-stent thrombus underwent initial stenting due to occlusive left pulmonary artery thrombus acutely post-superior cavopulmonary connection. There were seven (22%) significant bleeding complications for advanced anticoagulation and none for aspirin (p < 0.001). CONCLUSIONS Antithrombotic strategy does not appear to affect rates of in-stent thrombus in single-ventricle circulations. Aspirin alone may be sufficient for most patients undergoing stent implantation, while pre-existing thrombus may warrant advanced anticoagulation.
Collapse
|
5
|
Abstract
Background: Calculation of fractional flow reserve (FFR) using computed tomography (CT)-based 3D anatomical models and computational fluid dynamics (CFD) has become a common method to non-invasively assess the functional severity of atherosclerotic narrowing in coronary arteries. We examined the impact of various inflow boundary conditions on computation of FFR to shed light on the requirements for inflow boundary conditions to ensure model representation. Methods: Three-dimensional anatomical models of coronary arteries for four patients with mild to severe stenosis were reconstructed from CT images. FFR and its commonly-used alternatives were derived using the models and CFD. A combination of four types of inflow boundary conditions (BC) was employed: pulsatile, steady, patient-specific and population average. Results: The maximum difference of FFR between pulsatile and steady inflow conditions was 0.02 (2.4%), approximately at a level similar to a reported uncertainty level of clinical FFR measurement (3–4%). The flow with steady BC appeared to represent well the diastolic phase of pulsatile flow, where FFR is measured. Though the difference between patient-specific and population average BCs affected the flow more, the maximum discrepancy of FFR was 0.07 (8.3%), despite the patient-specific inflow of one patient being nearly twice as the population average. Conclusions: In the patients investigated, the type of inflow boundary condition, especially flow pulsatility, does not have a significant impact on computed FFRs in narrowed coronary arteries.
Collapse
|
6
|
Al Ghaithi R, Mori J, Nagy Z, Maclachlan A, Hardy L, Philippou H, Hethershaw E, Morgan NV, Senis YA, Harrison P. Evaluation of the Total Thrombus-Formation System (T-TAS): application to human and mouse blood analysis. Platelets 2018; 30:893-900. [PMID: 30365350 DOI: 10.1080/09537104.2018.1535704] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 12/26/2022]
Abstract
The Total Thrombus-formation Analyser System (T-TAS) is a whole blood flow chamber system for the measurement of in vitro thrombus formation under variable shear stress conditions. Our current study sought to evaluate the potential utility of the T-TAS for the measurement of thrombus formation within human and mouse whole blood. T-TAS microchips (collagen, PL chip; collagen/tissue thromboplastin, AR chip) were used to analyze platelet (PL) or fibrin-rich thrombus formation, respectively. Blood samples from humans (healthy and patients with mild bleeding disorders) and wild-type (WT), mice were tested. Light transmission lumi-aggregometer (lumi-LTA) was performed in PRP using several concentrations of ADP, adrenaline, arachidonic acid, collagen, PAR-1 peptide and ristocetin. Thrombus growth (N = 22) increased with shear within PL (4:40 ± 1.11, 3:25 ± 0.43 and 3:12 ± 0.48 mins [1000, 1500 and 2000s-1]) and AR chips (3:55 ± 0.42 and 1:49 ± 0.19 [240s-1 and 600s-1]). The area under the curve (AUC) on the PL chip was also reduced at 1000s-1 compared to 1500/2000s-1 (260 ± 51.7, 317 ± 55.4 and 301 ± 66.2, respectively). In contrast, no differences in the AUC between 240s-1 and 600s-1 were observed in the AR chip (1593 ± 122 and 1591 ± 158). The intra-assay coefficient of variation (CV) (n = 10) in the PL chip (1000s-1) and AR chip (240s-1) were T1014.1%, T6016.7%, T10-6022.8% and AUC1024.4% or T10 9.03%, T808.64%, T10-8023.8% and AUC305.1%. AR chip thrombus formation was inhibited by rivaroxaban (1 µM), but not with ticagrelor (10 µM). In contrast, PL chip thrombus formation was totally inhibited by ticagrelor. T-TAS shows an overall agreement with lumi-LTA in 87% of patients (n = 30) with normal PL counts recruited into the genotyping and phenotyping of platelet (GAPP) study and suspected to have a PL function defect. The onset (T10) of thrombus formation in WT mice (N = 4) was shorter when compared to humans e.g. PL chip (1000s-1) T10 were 02:02 ± 00:23 and 03:30 ± 0:45, respectively). T-TAS measures in vitro thrombus formation and can be used for monitoring antithrombotic therapy, investigating patients with suspected PL function defects and monitoring PL function within mice.
Collapse
Affiliation(s)
- Rashid Al Ghaithi
- Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, University of Birmingham , Birmingham , UK
- Haematology and Blood Transfusion Department, The Royal Hospital, Ministry of Health , Muscat , Sultanate of Oman
| | - Jun Mori
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, University of Birmingham , Birmingham , UK
| | - Zoltan Nagy
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, University of Birmingham , Birmingham , UK
| | - Annabel Maclachlan
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, University of Birmingham , Birmingham , UK
| | - Lewis Hardy
- Leeds Institute of Cardiovascular and Metabolic Medicine LIGHT Laboratories, University of Leeds , Leeds , West Yorkshire , UK
| | - Helen Philippou
- Leeds Institute of Cardiovascular and Metabolic Medicine LIGHT Laboratories, University of Leeds , Leeds , West Yorkshire , UK
| | - Emma Hethershaw
- Leeds Institute of Cardiovascular and Metabolic Medicine LIGHT Laboratories, University of Leeds , Leeds , West Yorkshire , UK
| | - Neil V Morgan
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, University of Birmingham , Birmingham , UK
| | - Yotis A Senis
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, University of Birmingham , Birmingham , UK
| | - Paul Harrison
- Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| |
Collapse
|
7
|
Ng J, Bourantas CV, Torii R, Ang HY, Tenekecioglu E, Serruys PW, Foin N. Local Hemodynamic Forces After Stenting: Implications on Restenosis and Thrombosis. Arterioscler Thromb Vasc Biol 2017; 37:2231-2242. [PMID: 29122816 DOI: 10.1161/atvbaha.117.309728] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/13/2017] [Indexed: 12/19/2022]
Abstract
Local hemodynamic forces are well-known to modulate atherosclerotic evolution, which remains one of the largest cause of death worldwide. Percutaneous coronary interventions with stent implantation restores blood flow to the downstream myocardium and is only limited by stent failure caused by restenosis, stent thrombosis, or neoatherosclerosis. Cumulative evidence has shown that local hemodynamic forces affect restenosis and the platelet activation process, modulating the pathophysiological mechanisms that lead to stent failure. This article first covers the pathophysiological mechanisms through which wall shear stress regulates arterial disease formation/neointima proliferation and the role of shear rate on stent thrombosis. Subsequently, the article reviews the current evidence on (1) the implications of stent design on the local hemodynamic forces, and (2) how stent/scaffold expansion can influence local flow, thereby affecting the risk of adverse events.
Collapse
Affiliation(s)
- Jaryl Ng
- From the National Heart Centre Singapore (J.N., H.Y.A., N.F.); Department of Biomedical Engineering, National University of Singapore, Singapore (J.N.); Departments of Cardiovascular Sciences (C.V.B.) and Mechanical Engineering (R.T.), University College London, United Kingdom; Department of Cardiology, Barts Health NHS Trust, London, United Kingdom (C.V.B.); Thoraxcenter, Erasmus MC, Rotterdam Erasmus University, The Netherlands (E.T., P.W.S.); National Heart & Lung Institute, Imperial College London, United Kingdom (P.W.S.); and Duke-NUS Medical School, National University of Singapore (N.F.)
| | - Christos V Bourantas
- From the National Heart Centre Singapore (J.N., H.Y.A., N.F.); Department of Biomedical Engineering, National University of Singapore, Singapore (J.N.); Departments of Cardiovascular Sciences (C.V.B.) and Mechanical Engineering (R.T.), University College London, United Kingdom; Department of Cardiology, Barts Health NHS Trust, London, United Kingdom (C.V.B.); Thoraxcenter, Erasmus MC, Rotterdam Erasmus University, The Netherlands (E.T., P.W.S.); National Heart & Lung Institute, Imperial College London, United Kingdom (P.W.S.); and Duke-NUS Medical School, National University of Singapore (N.F.)
| | - Ryo Torii
- From the National Heart Centre Singapore (J.N., H.Y.A., N.F.); Department of Biomedical Engineering, National University of Singapore, Singapore (J.N.); Departments of Cardiovascular Sciences (C.V.B.) and Mechanical Engineering (R.T.), University College London, United Kingdom; Department of Cardiology, Barts Health NHS Trust, London, United Kingdom (C.V.B.); Thoraxcenter, Erasmus MC, Rotterdam Erasmus University, The Netherlands (E.T., P.W.S.); National Heart & Lung Institute, Imperial College London, United Kingdom (P.W.S.); and Duke-NUS Medical School, National University of Singapore (N.F.)
| | - Hui Ying Ang
- From the National Heart Centre Singapore (J.N., H.Y.A., N.F.); Department of Biomedical Engineering, National University of Singapore, Singapore (J.N.); Departments of Cardiovascular Sciences (C.V.B.) and Mechanical Engineering (R.T.), University College London, United Kingdom; Department of Cardiology, Barts Health NHS Trust, London, United Kingdom (C.V.B.); Thoraxcenter, Erasmus MC, Rotterdam Erasmus University, The Netherlands (E.T., P.W.S.); National Heart & Lung Institute, Imperial College London, United Kingdom (P.W.S.); and Duke-NUS Medical School, National University of Singapore (N.F.)
| | - Erhan Tenekecioglu
- From the National Heart Centre Singapore (J.N., H.Y.A., N.F.); Department of Biomedical Engineering, National University of Singapore, Singapore (J.N.); Departments of Cardiovascular Sciences (C.V.B.) and Mechanical Engineering (R.T.), University College London, United Kingdom; Department of Cardiology, Barts Health NHS Trust, London, United Kingdom (C.V.B.); Thoraxcenter, Erasmus MC, Rotterdam Erasmus University, The Netherlands (E.T., P.W.S.); National Heart & Lung Institute, Imperial College London, United Kingdom (P.W.S.); and Duke-NUS Medical School, National University of Singapore (N.F.)
| | - Patrick W Serruys
- From the National Heart Centre Singapore (J.N., H.Y.A., N.F.); Department of Biomedical Engineering, National University of Singapore, Singapore (J.N.); Departments of Cardiovascular Sciences (C.V.B.) and Mechanical Engineering (R.T.), University College London, United Kingdom; Department of Cardiology, Barts Health NHS Trust, London, United Kingdom (C.V.B.); Thoraxcenter, Erasmus MC, Rotterdam Erasmus University, The Netherlands (E.T., P.W.S.); National Heart & Lung Institute, Imperial College London, United Kingdom (P.W.S.); and Duke-NUS Medical School, National University of Singapore (N.F.)
| | - Nicolas Foin
- From the National Heart Centre Singapore (J.N., H.Y.A., N.F.); Department of Biomedical Engineering, National University of Singapore, Singapore (J.N.); Departments of Cardiovascular Sciences (C.V.B.) and Mechanical Engineering (R.T.), University College London, United Kingdom; Department of Cardiology, Barts Health NHS Trust, London, United Kingdom (C.V.B.); Thoraxcenter, Erasmus MC, Rotterdam Erasmus University, The Netherlands (E.T., P.W.S.); National Heart & Lung Institute, Imperial College London, United Kingdom (P.W.S.); and Duke-NUS Medical School, National University of Singapore (N.F.).
| |
Collapse
|
8
|
Plasminogen activator inhibitor type 1 in platelets induces thrombogenicity by increasing thrombolysis resistance under shear stress in an in-vitro flow chamber model. Thromb Res 2016; 146:69-75. [PMID: 27611498 DOI: 10.1016/j.thromres.2016.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/26/2016] [Accepted: 09/02/2016] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Despite the proven benefits of thrombolytic therapy with tissue plasminogen activator (t-PA) for peripheral thromboembolism, perfusion failure frequently occurs, particularly in arterial circulation. We evaluated how the modification of fibrinolytic activity affects thrombus formation under flow and static conditions. MATERIALS AND METHODS t-PA-treated human whole-blood samples (n=6) were perfused over a microchip coated with collagen and tissue thromboplastin at different shear rates, and thrombus formation was quantified by measuring flow pressure changes. For comparison, rotational thromboelastometry (ROTEM) was used to evaluate fibrinolytic activity under static conditions. RESULTS At a shear rate of 240s-1, t-PA (200-800IU/ml) concentration-dependently delayed capillary occlusion, whereas at 600s-1, capillary occlusion was significantly faster and t-PA had limited effects, even at a supra-pharmacological concentration (800IU/ml). In contrast, 200IU/ml t-PA efficiently prevented clot formation in the ROTEM assay. The combined treatment of blood with a specific PAI-1 inhibitor (PAI-039) moderately enhanced the efficacy of t-PA, but only under flow conditions. In addition, 1:1-diluted blood samples of PAI-1-deficient (-/-) mice showed a significant delay of capillary occlusion at 240s-1, compared with those from wild-type mice (1.55 fold; P<0.001). This delayed occlusion was reproduced in samples containing platelets from PAI-1-/- and plasma from wild type, but was not observed by the opposite combination of blood components. CONCLUSIONS The present results suggest that the anti-thrombotic efficacy of t-PA is sensitive to arterial shear flow, and that PAI-1 secreted from activated platelets plays an essential role in thrombolytic resistance.
Collapse
|
9
|
Glynn JJ, Hinds MT. Bioactive Anti-Thrombotic Modification of Decellularized Matrix for Vascular Applications. Adv Healthc Mater 2016; 5:1439-46. [PMID: 27072858 PMCID: PMC5753589 DOI: 10.1002/adhm.201600020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 02/04/2016] [Indexed: 12/20/2022]
Abstract
The decellularized matrix derived from porcine small intestinal submucosa (SIS) is a widely used biomaterial being investigated for numerous applications. Currently, thrombus deposition and neointimal hyperplasia have limited the use of SIS in some vascular applications. To limit these detrimental processes, this work applies bioactive, endothelial-inspired properties to the material. SIS is modified with the endothelial cell membrane protein thrombomodulin and the glycosaminoglycan heparin to facilitate protein C activation and anticoagulant activity, respectively. Modifying SIS with thrombomodulin alone enables robust activated protein C (APC) generation, and thrombomodulin activity is maintained after prolonged exposure to fluid shear and blood plasma. Heparin-modified SIS has a potent anticoagulant activity. When both modifications are applied sequentially, SIS modified first with thrombomodulin then with heparin retains the full activity of each individual modification. Tubular SIS devices are connected to a baboon arteriovenous shunt to quantify thrombus deposition on these materials. After being exposed to flowing whole blood for 60 min, SIS devices modified first with thrombomodulin then with heparin have significantly less platelet accumulation compared to unmodified SIS devices. These studies demonstrate that modifying SIS with thrombomodulin and heparin confers APC generation and anticoagulant activity that results in reduced thrombogenesis.
Collapse
Affiliation(s)
- Jeremy J Glynn
- Department of Biomedical Engineering, Oregon Health & Science University, Mail Code: CH13B 3303 SW Bond Ave, Portland, OR, 97239, USA
| | - Monica T Hinds
- Department of Biomedical Engineering, Oregon Health & Science University, Mail Code: CH13B 3303 SW Bond Ave, Portland, OR, 97239, USA
| |
Collapse
|
10
|
Platelet function and coagulation in patients with STEMI and peri-interventional clopidogrel plus heparin vs. prasugrel plus bivalirudin therapy (BRAVE 4 substudy). Thromb Res 2015; 137:72-78. [PMID: 26639204 DOI: 10.1016/j.thromres.2015.11.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/24/2015] [Accepted: 11/14/2015] [Indexed: 11/23/2022]
Abstract
INTRODUCTION In this prespecified BRAVE 4 substudy we examined the antiplatelet and anticoagulant efficacy of clopidogrel plus heparin vs. prasugrel plus bivalirudin in patients with ST-segment elevation myocardial infarction. METHODS 26 patients received clopidogrel/heparin, 25 patients received prasugrel/bivalirudin and 20 additional untreated patients served as controls. Platelet aggregation was tested using whole blood impedance aggregometry. Dynamic platelet adhesion and aggregate formation to collagen were quantified under flow conditions. Coagulation tests included activated partial thromboplastin time (aPTT), international normalized ratio (INR) as well as rotational thrombelastography (ROTEM®). Analyses were performed 3 and 72h after drug administration. RESULTS At 3, but not at 72h we observed a significant increase in the inhibition of platelet aggregation in response to adenosine diphosphate (P<0.01), but not to arachidonic acid, collagen or thrombin receptor agonist in the prasugrel/bivalirudin group compared to the clopidogrel/heparin group. Inhibition of platelet adhesion to collagen under flow was significantly stronger in the prasugrel/bivalirudin group at 3 and 72h after drug administration (P<0.01). APTT was significantly higher in the clopidogrel/heparin group (P<0.05) and INR was significantly higher in the prasugrel/bivalirudin group (P<0.01) 3h after drug administration. Concerning ROTEM® analysis the drug combinations did not differ in reducing clot formation time (CFT) and both combinations did not influence maximum clot firmness (MCF) compared to the controls. CONCLUSIONS We could demonstrate a more pronounced inhibition of platelet aggregation as well as platelet adhesion and aggregate formation to collagen under flow in prasugrel plus bivalirudin treated patients.
Collapse
|
11
|
|
12
|
Momtahan N, Sukavaneshvar S, Roeder BL, Cook AD. Strategies and processes to decellularize and recellularize hearts to generate functional organs and reduce the risk of thrombosis. TISSUE ENGINEERING PART B-REVIEWS 2014; 21:115-32. [PMID: 25084164 DOI: 10.1089/ten.teb.2014.0192] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Heart failure is one of the leading causes of death in the United States. Current therapies, such as heart transplants and bioartificial hearts, are helpful, but not optimal. Decellularization of porcine whole hearts followed by recellularization with patient-specific human cells may provide the ultimate solution for patients with heart failure. Great progress has been made in the development of efficient processes for decellularization, and the design of automated bioreactors. Challenges remain in selecting and culturing cells, growing the cells on the decellularized scaffolds without contamination, characterizing the regenerated organs, and preventing thrombosis. Various strategies have been proposed to prevent thrombosis of blood-contacting devices, including reendothelization and the creation of nonfouling surfaces using surface modification technologies. This review discusses the progress and remaining challenges involved with recellularizing whole hearts, focusing on the prevention of thrombosis.
Collapse
Affiliation(s)
- Nima Momtahan
- 1 Department of Chemical Engineering, Brigham Young University , Provo, Utah
| | | | | | | |
Collapse
|
13
|
Foin N, Gutiérrez-Chico JL, Nakatani S, Torii R, Bourantas CV, Sen S, Nijjer S, Petraco R, Kousera C, Ghione M, Onuma Y, Garcia-Garcia HM, Francis DP, Wong P, Di Mario C, Davies JE, Serruys PW. Incomplete Stent Apposition Causes High Shear Flow Disturbances and Delay in Neointimal Coverage as a Function of Strut to Wall Detachment Distance. Circ Cardiovasc Interv 2014; 7:180-9. [DOI: 10.1161/circinterventions.113.000931] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Nicolas Foin
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Juan Luis Gutiérrez-Chico
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Shimpei Nakatani
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Ryo Torii
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Christos V. Bourantas
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Sayan Sen
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Sukhjinder Nijjer
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Ricardo Petraco
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Chrysa Kousera
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Matteo Ghione
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Yoshinobu Onuma
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Hector M. Garcia-Garcia
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Darrel P. Francis
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Philip Wong
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Carlo Di Mario
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Justin E. Davies
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| | - Patrick W. Serruys
- From the International Centre for Circulatory Health, NHLI, Imperial College London, London, United Kingdom (N.F., S.S., S.N., R.P., C.K., D.P.F., J.E.D., P.W.S.); National Heart Centre Singapore, Singapore (N.F., P.W.); ThoraxCenter, Erasmus Medical Center, Rotterdam, The Netherlands (J.L.G.-C., S.N., C.V.B., Y.O., H.M.G.-G., P.W.S.); Ludwig-Maximilian Universität, München, Germany (J.L.G.-C.); Department of Mechanical Engineering, University College London, London, United Kingdom (R.T.); NIHR, BRU
| |
Collapse
|
14
|
Li M, Hotaling NA, Ku DN, Forest CR. Microfluidic thrombosis under multiple shear rates and antiplatelet therapy doses. PLoS One 2014; 9:e82493. [PMID: 24404131 PMCID: PMC3880267 DOI: 10.1371/journal.pone.0082493] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 10/23/2013] [Indexed: 12/04/2022] Open
Abstract
The mainstay of treatment for thrombosis, the formation of occlusive platelet aggregates that often lead to heart attack and stroke, is antiplatelet therapy. Antiplatelet therapy dosing and resistance are poorly understood, leading to potential incorrect and ineffective dosing. Shear rate is also suspected to play a major role in thrombosis, but instrumentation to measure its influence has been limited by flow conditions, agonist use, and non-systematic and/or non-quantitative studies. In this work we measured occlusion times and thrombus detachment for a range of initial shear rates (500, 1500, 4000, and 10000 s(-1)) and therapy concentrations (0-2.4 µM for eptifibatide, 0-2 mM for acetyl-salicylic acid (ASA), 3.5-40 Units/L for heparin) using a microfluidic device. We also measured complete blood counts (CBC) and platelet activity using whole blood impedance aggregometry. Effects of shear rate and dose were analyzed using general linear models, logistic regressions, and Cox proportional hazards models. Shear rates have significant effects on thrombosis/dose-response curves for all tested therapies. ASA has little effect on high shear occlusion times, even at very high doses (up to 20 times the recommended dose). Under ASA therapy, thrombi formed at high shear rates were 4 times more prone to detachment compared to those formed under control conditions. Eptifibatide reduced occlusion when controlling for shear rate and its efficacy increased with dose concentration. In contrast, the hazard of occlusion from ASA was several orders of magnitude higher than that of eptifibatide. Our results show similar dose efficacy to our low shear measurements using whole blood aggregometry. This quantitative and statistically validated study of the effects of a wide range of shear rate and antiplatelet therapy doses on occlusive thrombosis contributes to more accurate understanding of thrombosis and to models for optimizing patient treatment.
Collapse
Affiliation(s)
- Melissa Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Nathan A Hotaling
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - David N Ku
- George W. Woodruff Department of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Craig R Forest
- George W. Woodruff Department of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| |
Collapse
|
15
|
Qu Z, Krishnamurthy V, Haller CA, Dorr BM, Marzec UM, Hurst S, Hinds MT, Hanson SR, Liu DR, Chaikof EL. Immobilization of actively thromboresistant assemblies on sterile blood-contacting surfaces. Adv Healthc Mater 2014; 3:30-5. [PMID: 23788402 DOI: 10.1002/adhm.201300110] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Indexed: 12/13/2022]
Abstract
Rapid one-step modification of thrombomodulin with alkylamine derivatives such as azide, biotin, and PEG is achieved using an evolved sortase (eSrtA) mutant. The feasibility of a point-of-care scheme is demonstrated herein to site-specifically immobilize azido-thrombomodulin on sterilized commercial ePTFE vascular grafts, which exhibit superior thromboresistance compared with commercial heparin-coated grafts in a primate model of acute graft thrombosis.
Collapse
Affiliation(s)
- Zheng Qu
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School and the Wyss Institute of Biologically Inspired, Engineering of Harvard University, Boston, MA 02115, USA; Coulter Department of Biomedical Engineering, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Hosokawa K, Ohnishi T, Miura N, Sameshima H, Koide T, Tanaka KA, Maruyama I. Antithrombotic effects of PAR1 and PAR4 antagonists evaluated under flow and static conditions. Thromb Res 2014; 133:66-72. [DOI: 10.1016/j.thromres.2013.10.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/09/2013] [Accepted: 10/28/2013] [Indexed: 01/22/2023]
|
17
|
Ferkau A, Ecklebe S, Jahn K, Calmer S, Theilmeier G, Mischke R. A dynamic flow-chamber-based adhesion assay to assess canine platelet-matrix interactions in vitro. Vet Clin Pathol 2013; 42:150-6. [DOI: 10.1111/vcp.12035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Annika Ferkau
- Department of Anesthesiology and Intensive Care Medicine; Hannover Medical School; Hannover; Germany
| | - Silke Ecklebe
- Department of Anesthesiology and Intensive Care Medicine; Hannover Medical School; Hannover; Germany
| | - Kira Jahn
- Department of Anesthesiology and Intensive Care Medicine; Hannover Medical School; Hannover; Germany
| | - Simone Calmer
- Department of Anesthesiology and Intensive Care Medicine; Hannover Medical School; Hannover; Germany
| | - Gregor Theilmeier
- Department of Anesthesiology and Intensive Care Medicine; Hannover Medical School; Hannover; Germany
| | - Reinhard Mischke
- Small Animal Clinic; University of Veterinary Medicine; Hannover; Germany
| |
Collapse
|
18
|
Evaluation of Blood–Materials Interactions. Biomater Sci 2013. [DOI: 10.1016/b978-0-08-087780-8.00055-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
|
19
|
Ngo DH, Vo TS, Ngo DN, Wijesekara I, Kim SK. Biological activities and potential health benefits of bioactive peptides derived from marine organisms. Int J Biol Macromol 2012; 51:378-83. [DOI: 10.1016/j.ijbiomac.2012.06.001] [Citation(s) in RCA: 200] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 06/01/2012] [Indexed: 01/17/2023]
|
20
|
Joglekar MV, Ware J, Xu J, Fitzgerald MEC, Gartner TK. Platelets, glycoprotein Ib-IX, and von Willebrand factor are required for FeCl(3)-induced occlusive thrombus formation in the inferior vena cava of mice. Platelets 2012; 24:205-12. [PMID: 22720736 DOI: 10.3109/09537104.2012.696746] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Venous thromboembolism is a leading cause of death from cardiovascular disease. Despite the importance of the glycoprotein (GP) Ib-IX/von Willebrand factor (vWF) axis in arterial thrombosis, its requirement in venous, not venule thrombosis in response to endothelial injury (not stenosis or stasis) is uncharacterized. GPIbα-vWF participation in FeCl(3)-induced thrombus formation was evaluated in the inferior vena cava (IVC). Stable, occlusive thrombus formation in response to FeCl(3)-induced injury of the IVC was studied. FeCl(3) (20% FeCl(3), 10 minutes)-induced occlusive thrombosis required platelets as confirmed by a lack of occlusion in thrombocytopenic mice, and stable occlusion in control animals. No IVC occlusion was observed using GPIbα-deficient animals, a model of the human Bernard-Soulier syndrome (BSS). Transgenic IL-4 R/GPIbα mice (lack murine GPIbα, but express the extracellular domain of the human interleukin (IL-4 receptor fused to the transmembrane and cytoplasmic domains of human GPIbα) were studied to determine if the absence of IVC occlusion in the BSS mouse was caused by GPIbα extracellular domain deficiency rather than platelet BSS phenotype associated abnormalities. As with GPIbα knock-out mice, no occlusion was observed in the IVC of IL-4 R/GPIbα mice. The IVC of vWF-deficient mice also failed to occlude in response to FeCl(3) treatment. The chimeric protein GPIbα(2V)-Fc prevented occlusion, demonstrating that GPIbα-vWF A1 domain interaction is required for FeCl(3)-induced stable thrombus formation in the IVC. Therefore, FeCl(3)-induced stable, occlusive thrombus formation in the IVC is platelet, and apparently GPIbα-vWF interaction dependent, despite the large diameter and low venous flow rate in the IVC.
Collapse
Affiliation(s)
- M V Joglekar
- Division of Hematology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
21
|
Ogawa S, Szlam F, Dunn AL, Bolliger D, Ohnishi T, Hosokawa K, Tanaka KA. Evaluation of a novel flow chamber system to assess clot formation in factor VIII-deficient mouse and anti-factor IXa-treated human blood. Haemophilia 2012; 18:926-32. [PMID: 22642581 DOI: 10.1111/j.1365-2516.2012.02867.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2012] [Indexed: 11/30/2022]
Abstract
Blood flow properties play important roles in the regulation and formation of thrombus. To evaluate the influence of blood flow on thrombus formation in haemophilia, whole blood samples were obtained from FVIII-deficient (FVIII(-/-) ) and wild-type (FVIII(+/+) ) mice (n = 6 respectively), and from six human volunteers. Anti-FIXa aptamer was added to human blood to model acquired haemophilia B. Recalcified whole blood samples containing corn trypsin inhibitor and danaproid were perfused over the microchip coated with collagen and tissue thromboplastin at shear rates of 1100 and 110 s(-1) . Thrombus formation in the capillary was quantified by monitoring flow pressure changes. The intervals to 5 kPa (T(5) ) and 40 k Pa (T(40) ) reflect the onset and growth of thrombus formation respectively. Furthermore, fibrin and platelets in thrombi were quantified by immunostaining. T(5) at both shear rates were similar in FVIII(-/-) and FVIII(+/+) mice. T(40) of FVIII(-/-) mice (1569 ± 565 s) was significantly delayed compared with FVIII(+/+) mice (339 ± 78 s) at 110 s(-1) (P < 0.05), but not at 1100 s(-1) . The delay was normalized by adding human FVIII (2 IU mL(-1) ). Similarly, adding anti-FIXa aptamer to human blood prolonged T(40) at 110 s(-1) (P < 0.01), but not at 1100 s(-1) . Impaired production of fibrin due to anti-FIXa aptamer at 110 s(-1) was shown in the immunostained thrombus. Our perfusion experiments demonstrated that shear rates influence thrombus formation patterns in haemophilia, and that reduced activity of intrinsic tenase (FIXa-FVIIIa) becomes evident under venous shear rates.
Collapse
Affiliation(s)
- S Ogawa
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Wolzt M, Eriksson UG, Gouya G, Leuchten N, Kapiotis S, Elg M, Schützer KM, Zetterstrand S, Holmberg M, Wåhlander K. Effect on perfusion chamber thrombus size in patients with atrial fibrillation during anticoagulant treatment with oral direct thrombin inhibitors, AZD0837 or ximelagatran, or with vitamin K antagonists. Thromb Res 2012; 129:e83-91. [DOI: 10.1016/j.thromres.2011.08.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 08/12/2011] [Accepted: 08/17/2011] [Indexed: 11/30/2022]
|
23
|
Qu Z, Muthukrishnan S, Urlam MK, Haller CA, Jordan SW, Kumar VA, Marzec UM, Elkasabi Y, Lahann J, Hanson SR, Chaikof EL. A biologically active surface enzyme assembly that attenuates thrombus formation. ADVANCED FUNCTIONAL MATERIALS 2011; 21:4736-4743. [PMID: 23532366 PMCID: PMC3606904 DOI: 10.1002/adfm.201101687] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Activation of hemostatic pathways by blood-contacting materials remains a major hurdle in the development of clinically durable artificial organs and implantable devices. We postulate that surface-induced thrombosis may be attenuated by the reconstitution onto blood contacting surfaces of bioactive enzymes that regulate the production of thrombin, a central mediator of both clotting and platelet activation cascades. Thrombomodulin (TM), a transmembrane protein expressed by endothelial cells, is an established negative regulator of thrombin generation in the circulatory system. Traditional techniques to covalently immobilize enzymes on solid supports may modify residues contained within or near the catalytic site, thus reducing the bioactivity of surface enzyme assemblies. In this report, we present a molecular engineering and bioorthogonal chemistry approach to site-specifically immobilize a biologically active recombinant human TM fragment onto the luminal surface of small diameter prosthetic vascular grafts. Bioactivity and biostability of TM modified grafts is confirmed in vitro and the capacity of modified grafts to reduce platelet activation is demonstrated using a non-human primate model. These studies indicate that molecularly engineered interfaces that display TM actively limit surface-induced thrombus formation.
Collapse
Affiliation(s)
- Zheng Qu
- Departments of Biomedical Engineering and Surgery, Georgia Institute of Technology and Emory University Atlanta, GA 30322 (USA)
| | - Sharmila Muthukrishnan
- Departments of Biomedical Engineering and Surgery, Georgia Institute of Technology and Emory University Atlanta, GA 30322 (USA)
| | - Murali K. Urlam
- Departments of Biomedical Engineering and Surgery, Georgia Institute of Technology and Emory University Atlanta, GA 30322 (USA)
| | - Carolyn A. Haller
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, and the Wyss Institute of Biologically Inspired Engineering of Harvard University Boston, MA 02115 (USA)
| | - Sumanas W. Jordan
- Departments of Biomedical Engineering and Surgery, Georgia Institute of Technology and Emory University Atlanta, GA 30322 (USA)
| | - Vivek A. Kumar
- Departments of Biomedical Engineering and Surgery, Georgia Institute of Technology and Emory University Atlanta, GA 30322 (USA)
| | - Ulla M. Marzec
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006 (USA)
| | - Yaseen Elkasabi
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 (USA)
| | - Joerg Lahann
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 (USA)
| | - Stephen R. Hanson
- Oregon National Primate Research Center, Oregon Health and Science University Beaverton, OR 97006 (USA)
| | - Elliot L. Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, and the Wyss Institute of Biologically Inspired Engineering of Harvard University Boston, MA 02115 (USA)
| |
Collapse
|
24
|
Hosokawa K, Ohnishi T, Fukasawa M, Kondo T, Sameshima H, Koide T, Tanaka KA, Maruyama I. A microchip flow-chamber system for quantitative assessment of the platelet thrombus formation process. Microvasc Res 2011; 83:154-61. [PMID: 22166857 DOI: 10.1016/j.mvr.2011.11.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 11/08/2011] [Accepted: 11/27/2011] [Indexed: 10/14/2022]
Abstract
As the pathogenesis of arterial thrombosis often includes platelet thrombus formation (PTF), antiplatelet agents are commonly used for the prevention of thromboembolic events. Here, using a novel microchip flow-chamber system we developed to quantitatively analyze the PTF process, we evaluated the pharmacological efficacies of antiplatelet agents under different arterial shear rates. Hirudin-anticoagulated whole blood was perfused over a collagen-coated microchip at shear rates of 1000, 1500, and 2000s(-1), and PTF in the absence and presence of various antiplatelet agents was observed microscopically and quantified by measuring flow-pressure changes. The onset of PTF was measured as T(10) (time to reach 10 kPa), and AUC(10) (area under the flow pressure curve for the first 10 min) was calculated to quantify the overall stability of the formed thrombus. Aspirin and AR-C66096 (P2Y(12)-antagonist) at high concentrations (50 μM and 1000 nM, respectively) prolonged T(10) only modestly (AR-C66096>aspirin), but effectively decreased AUC(10), resulting in unstable PTF at all examined shear rates. With dual inhibition using both aspirin (25 μM) and ARC-66096 (250 nM), AUC(10) was drastically reduced. Nearly complete suppression of AUC(10) was also observed with abciximab (2 μg ml(-1)) and beraprost (PGI(2)-analog; 4 nM). Although OS-1 (GPIbα-antagonist; 100 nM) prevented complete capillary occlusion, significant amounts of microscopic thrombi were observed on the collagen surface. In contrast to abciximab and beraprost, OS-1 differentially affected PTF under higher shear conditions. Our novel analytical system is capable of distinguishing the pharmacological effects of various antiplatelet agents under physiological shear rates, suggesting that this system may aid in the determination of the appropriate type and dose of antiplatelet agent in the clinical setting.
Collapse
Affiliation(s)
- Kazuya Hosokawa
- Department of System Biology in Thromboregulation, Kagoshima University, Kagoshima 890-8520, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Pugh N, Jarvis GE, Koch A, Sakariassen KS, Davis B, Farndale RW. The impact of factor Xa inhibition on axial dependent arterial thrombus formation triggered by a tissue factor rich surface. J Thromb Thrombolysis 2011; 33:6-15. [PMID: 22120925 DOI: 10.1007/s11239-011-0658-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
This study was designed to assess the effect of Factor Xa antagonists on thrombus formation at various axial positions on a tissue factor rich surface under arterial blood flow conditions. Non-anticoagulated, flowing human blood, drawn directly from an antecubital vein, was perfused over a tissue factor coated cover slip in a parallel-plate perfusion chamber. Thrombus surface coverage, thrombus mean height and fibrin surface coverage were measured at six different axial positions by confocal microscopy. Both thrombus surface coverage and mean height decreased along the cover slip axis whereas the fibrin surface coverage increased. Pre-chamber treatment of blood with the direct Factor Xa inhibitors Razaxaban and 813893 resulted in significantly reduced thrombus and fibrin formation at all axial positions investigated (P < 0.05). Thrombus and fibrin deposition in a laminar flow chamber changed with axial position with surface coverage measurements being more reproducible than thrombus mean height. Data were more reproducible towards the centre of the flow chamber than at the extremities. Razaxaban and 813893 inhibited thrombus and fibrin formation at the highest concentrations tested. No difference in drug effect was apparent at different axial positions. In conclusion, axial position influences the degree of thrombus and fibrin deposition with measurements being less reproducible at the extremities of the flow chamber. This technique may prove useful for analysing anti-thrombotic drug effects before progression to clinical trials.
Collapse
Affiliation(s)
- Nicholas Pugh
- Department of Biochemistry, University of Cambridge, Cambridge CB1 2QW, UK.
| | | | | | | | | | | |
Collapse
|
26
|
Hosokawa K, Ohnishi T, Kondo T, Fukasawa M, Koide T, Maruyama I, Tanaka KA. A novel automated microchip flow-chamber system to quantitatively evaluate thrombus formation and antithrombotic agents under blood flow conditions. J Thromb Haemost 2011; 9:2029-37. [PMID: 21827607 DOI: 10.1111/j.1538-7836.2011.04464.x] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND AIMS In the present study, we describe a newly developed microchip-based analytical system to evaluate white thrombus formation (WTF). Efficacies of various antithrombotic agents were compared under different flow conditions. METHODS Whole blood containing corn trypsin inhibitor was perfused over a microchip coated with collagen and tissue thromboplastin at the lower and higher shear rates of 240 and 600 s(-1) , and WTF process inside the microchip was quantified by monitoring a flow pressure. Parameters of T(10) (time to 10 kPa), T(10-80) (time from 10 to 80 kPa) and OT (occlusion time; time to 80 kPa) were used to evaluate the onset and the growth rate of WTF, and the capillary occlusion, respectively. RESULTS After perfusion was started, white thrombus composed of activated platelets and fibrin was formed on the coated surface. Thrombus gradually increased in size and eventually occluded the capillary. Among anticoagulants, heparin (0.5-1.0 U mL(-1)) potently prolonged T(10) at both shear rates, whereas low molecular weight heparin (1.0-2.0 IU mL(-1)) inhibited the growth of WTF at the lower shear rate. Among antiplatelet agents, abciximab (1-2 μg mL(-1)) significantly reduced the size and number of thrombi, which was additively enhanced in the presence of heparin (0.5 U mL(-1) ). OS-1 (specific GPIbα-antagonist) prevented the complete capillary occlusion. CONCLUSION The novel monitoring system of WTF may be useful in preclinical and clinical evaluations of different types of antithrombotic strategies, and their effects in combination.
Collapse
Affiliation(s)
- K Hosokawa
- Research Institute, Fujimori Kogyo Co., Yokohama, Kanagawa, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Rugonyi S. Effect of blood flow on near-the-wall mass transport of drugs and other bioactive agents: a simple formula to estimate boundary layer concentrations. J Biomech Eng 2008; 130:021010. [PMID: 18412497 DOI: 10.1115/1.2899571] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Transport of bioactive agents through the blood is essential for cardiovascular regulatory processes and drug delivery. Bioactive agents and other solutes infused into the blood through the wall of a blood vessel or released into the blood from an area in the vessel wall spread downstream of the infusion/release region and form a thin boundary layer in which solute concentration is higher than in the rest of the blood. Bioactive agents distributed along the vessel wall affect endothelial cells and regulate biological processes, such as thrombus formation, atherogenesis, and vascular remodeling. To calculate the concentration of solutes in the boundary layer, researchers have generally used numerical simulations. However, to investigate the effect of blood flow, infusion rate, and vessel geometry on the concentration of different solutes, many simulations are needed, leading to a time-consuming effort. In this paper, a relatively simple formula to quantify concentrations in a tube downstream of an infusion/release region is presented. Given known blood-flow rates, tube radius, solute diffusivity, and the length of the infusion region, this formula can be used to quickly estimate solute concentrations when infusion rates are known or to estimate infusion rates when solute concentrations at a point downstream of the infusion region are known. The developed formula is based on boundary layer theory and physical principles. The formula is an approximate solution of the advection-diffusion equations in the boundary layer region when solute concentration is small (dilute solution), infusion rate is modeled as a mass flux, and there is no transport of solute through the wall or chemical reactions downstream of the infusion region. Wall concentrations calculated using the formula developed in this paper were compared to the results from finite element models. Agreement between the results was within 10%. The developed formula could be used in experimental procedures to evaluate drug efficacy, in the design of drug-eluting stents, and to calculate rates of release of bioactive substances at active surfaces using downstream concentration measurements. In addition to being simple and fast to use, the formula gives accurate quantifications of concentrations and infusion rates under steady-state and oscillatory flow conditions, and therefore can be used to estimate boundary layer concentrations under physiological conditions.
Collapse
Affiliation(s)
- Sandra Rugonyi
- Biomedical Engineering Department, Oregon Health & Science University, 3303 SW Bond Avenue, Portland, OR 97239, USA.
| |
Collapse
|
28
|
Raymond J, Guilbert F, Metcalfe A, Salazkin I, Gevry G, Leblanc P, Weill A, Roy D. In Vivo Thrombogenicity of Embolic Protection Systems for Angioplasty and Stenting. Interv Neuroradiol 2007; 13:329-33. [DOI: 10.1177/159101990701300403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Accepted: 11/12/2007] [Indexed: 11/15/2022] Open
Abstract
Despite the increasing use of embolic protection systems (EPS) for carotid stenting, their intrinsic in vivo thrombogenicity remains unknown. We studied three different types of EPS (n = 24) deployed in the carotid arteries of pigs in which pools of platelets and fibrinogen were labelled with 111In and 125I. The amount of clot deposition seen on photography was also scored using a qualitative scale. EPS made of fabric nets under normal flow conditions were 5–6 and 15–16 times more thrombogenic (for both platelet (P=.04) and fibrin (P=.007)) than Nitinol mesh nets. Clot deposition on Nitinol mesh nets was more abundant under flow arrest than under normal flow conditions (P=.018). EPS differ in intrinsic thrombogenicity, a characteristic of the material that could be investigated in pre-clinical studies designed to optimize devices.
Collapse
Affiliation(s)
- J. Raymond
- Radiology Department and Research Centre, CHUM — Notre-Dame Hospital, Montréal, Québec, CANADA
| | - F. Guilbert
- Radiology Department and Research Centre, CHUM — Notre-Dame Hospital, Montréal, Québec, CANADA
| | - A. Metcalfe
- Radiology Department and Research Centre, CHUM — Notre-Dame Hospital, Montréal, Québec, CANADA
| | - I. Salazkin
- Radiology Department and Research Centre, CHUM — Notre-Dame Hospital, Montréal, Québec, CANADA
| | - G. Gevry
- Radiology Department and Research Centre, CHUM — Notre-Dame Hospital, Montréal, Québec, CANADA
| | - P. Leblanc
- Radiology Department and Research Centre, CHUM — Notre-Dame Hospital, Montréal, Québec, CANADA
| | - A. Weill
- Radiology Department and Research Centre, CHUM — Notre-Dame Hospital, Montréal, Québec, CANADA
| | - D. Roy
- Radiology Department and Research Centre, CHUM — Notre-Dame Hospital, Montréal, Québec, CANADA
| |
Collapse
|
29
|
Sakariassen KS. Blood flow devices in medical research and clinical testing in humans: are we approaching personalized medicine? Future Cardiol 2007; 3:71-90. [DOI: 10.2217/14796678.3.1.71] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This review focuses on studies of blood flow devices employed in man to unravel the mechanisms of bleeding and thrombotic disorders, and on the characterization of novel experimental antithrombotic entities and drug candidates in biopharmaceutical research and development. Clinical studies with drug candidates and new therapeutic strategies have also been performed, and the predictability of these experimental approaches to clinical situations is excellent. Based on the solid validation of these flow devices, miniature flow devices employing nonanticoagulated blood drawn directly from an antecubital vein should be developed for diagnostic purposes. It is anticipated that such a diagnostic flow device could develop into a personalized medicine approach.
Collapse
|
30
|
Yamauchi F, Koyamatsu Y, Kato K, Iwata H. Layer-by-layer assembly of cationic lipid and plasmid DNA onto gold surface for stent-assisted gene transfer. Biomaterials 2006; 27:3497-504. [PMID: 16488471 DOI: 10.1016/j.biomaterials.2006.02.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Accepted: 02/02/2006] [Indexed: 11/21/2022]
Abstract
Intravascular stent-assisted gene transfer is an advanced approach for the therapy of vascular diseases such as atherosclerosis and stenosis. This approach requires a stent that allows local and efficient administration of therapeutic genes to the target cells at the vascular wall. To create such a stent, a method was developed for loading plasmid DNA onto the metal surface. The method involves the formation of self-assembled monolayer on the noble metal surface followed by electrostatic layer-by-layer (LBL) assembly of a cationic lipid/plasmid DNA complex and free plasmid DNA. In this in vitro feasibility study, the thin plainer film and the wire of gold were used as a substrate. The LBL assembly process was characterized by surface plasmon resonance spectroscopy and static contact angle measurement. Plasmid DNA loaded in the multilayer exhibited improved resistance against nuclease digestion. When cultured directly on the DNA-loaded surface, cells were transfected to express exogenous gene in the DNA loading-dependent manner. Plasmid DNA could also be transferred to endothelial cells from its apical side by placing the DNA-loaded gold wire onto the cell layer.
Collapse
Affiliation(s)
- Fumio Yamauchi
- Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | |
Collapse
|
31
|
Frojmovic M, Labarthe B, Legrand C. Inhibition and reversal of platelet aggregation by alphaIIbbeta3 antagonists depends on the anticoagulant and flow conditions: differential effects of Abciximab and Lamifiban. Br J Haematol 2006; 131:348-55. [PMID: 16225655 DOI: 10.1111/j.1365-2141.2005.05782.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Shear influences platelet aggregate formation and stability, as well as the inhibitory capacities of antithrombotic drugs. We compared the inhibitory and disaggregating properties of two distinct alphaIIbbeta3 antagonists, Abciximab and Lamifiban, on platelet aggregation induced by adenosine diphosphate (ADP) (5 micromol/l) in platelet-rich plasma (PRP), in an aggregometer (poorly defined low shear, <100/s) and in a microcouette at arterial shear rate (1,000/s). Platelet aggregation was detected by changes in light transmission in the aggregometer (TA), and by particle counting with a flow cytometer (PA). Lamifiban (1 mumol/l) completely inhibited TA or PA induced by ADP in citrated PRP in the aggregometer or microcouette. In contrast, Abciximab (2 micromol/l) only partially inhibited PA in the microcouette while blocking both TA and PA in the aggregometer. Moreover, Abciximab did not reverse platelet aggregates formed either in the microcouette or in the aggregometer, whereas Lamifiban caused complete reversal. On the contrary, Abciximab completely inhibited platelet aggregation induced by ADP in hirudin/d-Phe-Pro-Arg-chloromethylketone PRP in the microcouette. Our results demonstrate a marked dependence of inhibitory capacity of Abciximab on shear conditions, with citrate anticoagulant responsible for the residual aggregation, in contrast to Lamifiban, another alphaIIbbeta3 antagonist interacting with a distinct site on beta3.
Collapse
Affiliation(s)
- Mony Frojmovic
- INSERM U553, Institut Universitaire d'Hématologie Paris VII, Hôpital Saint-Louis, Paris, France
| | | | | |
Collapse
|
32
|
Cadroy Y, Thalamas C, Sakariassen K, Boneu B. Superior efficacy of clopidogrel plus acetylsalicylic acid compared with extended-release dipyridamole plus acetylsalicylic acid in preventing arterial thrombogenesis in healthy volunteers. Thromb Res 2005; 116:293-300. [PMID: 16038713 DOI: 10.1016/j.thromres.2004.12.014] [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] [Received: 11/22/2004] [Revised: 12/16/2004] [Accepted: 12/16/2004] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Recent ex vivo platelet aggregometry data indicate that clopidogrel 75 mg/day plus acetylsalicylic acid (ASA) 75 mg/day is a more potent antiplatelet regimen than the marketed combination of dipyridamole+ASA. The present study was designed to assess the antithrombotic effect of both dual antiplatelet regimens using a human ex vivo model of arterial thrombosis. MATERIALS AND METHODS This was a randomized, double-blind, placebo-controlled, crossover study. During two 10-day treatment periods separated by a 14-day washout period, 23 healthy male volunteers received once-daily clopidogrel 75 mg plus acetylsalicylic acid 75 mg, or twice-daily extended-release dipyridamole 200 mg plus acetylsalicylic acid 25 mg. Assessments were made at baseline and on Day 10 of each period. Arterial thrombus formation was induced ex vivo by exposing a collagen-coated surface in a parallel-plate perfusion chamber to native blood for 3 min (arterial wall shear rate 2600 s(-1)). Total platelet and fibrin deposition was determined by immunoenzymatic methods. RESULTS Compared with baseline values, the mean inhibition of total platelet deposition was 63.9+/-5.9% with clopidogrel plus acetylsalicylic acid, compared with 18.4+/-5.6% for extended-release dipyridamole plus acetylsalicylic acid (67% reduction; 95% CI, 49-79%; p<0.0001). Corresponding figures for fibrin deposition were 64.9+/-4.8% and 18.3+/-9.7%, respectively (58% reduction; 95% CI, 45-67%; p<0.0001). Both treatments were well tolerated. CONCLUSIONS Clopidogrel plus acetylsalicylic acid showed significantly superior antithrombotic efficacy compared with extended-release dipyridamole plus acetylsalicylic acid in preventing arterial thrombogenesis in humans.
Collapse
Affiliation(s)
- Yves Cadroy
- Laboratoire de Recherche sur l'Hémostase et la Thrombose, EA2049, CHU Rangueil, TSA 50032, 31059 Toulouse Cedex 9, France.
| | | | | | | |
Collapse
|
33
|
Gorbet MB, Sefton MV. Biomaterial-associated thrombosis: roles of coagulation factors, complement, platelets and leukocytes. Biomaterials 2005; 25:5681-703. [PMID: 15147815 DOI: 10.1016/j.biomaterials.2004.01.023] [Citation(s) in RCA: 857] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2003] [Accepted: 01/19/2004] [Indexed: 01/18/2023]
Abstract
Our failure to produce truly non-thrombogenic materials may reflect a failure to fully understand the mechanisms of biomaterial-associated thrombosis. The community has focused on minimizing coagulation or minimizing platelet adhesion and activation. We have infrequently considered the interactions between the two although we are generally familiar with these interactions. However, we have rarely considered in the context of biomaterial-associated thrombosis the other major players in blood: complement and leukocytes. Biomaterials are known agonists of complement and leukocyte activation, but this is frequently studied only in the context of inflammation. For us, thrombosis is a special case of inflammation. Here we summarize current perspectives on all four of these components in thrombosis and with biomaterials and cardiovascular devices. We also briefly highlight a few features of biomaterial-associated thrombosis that are not often considered in the biomaterials literature: The importance of tissue factor and the extrinsic coagulation system. Complement activation as a prelude to platelet activation and its role in thrombosis. The role of leukocytes in thrombin formation. The differing time scales of these contributions.
Collapse
Affiliation(s)
- Maud B Gorbet
- Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, 4 Taddle Creek Road, Room 407D, Toronto, Ont., Canada M5S 3G9
| | | |
Collapse
|
34
|
Levy Y, Shenkman B, Tamarin I, Pauzner R, Shoenfeld Y, Langevitz P, Savion N, Varon D. Increased platelet deposition on extracellular matrix under flow conditions in patients with antiphospholipid syndrome who experience thrombotic events. ACTA ACUST UNITED AC 2005; 52:4011-7. [PMID: 16320349 DOI: 10.1002/art.21437] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To assess platelet function under defined flow conditions in patients with antiphospholipid syndrome (APS) and to correlate the results with thrombotic complications and the presence of subsets of antiphospholipid antibodies (aPL), lupus anticoagulant (LAC), and/or anticardiolipin antibodies (aCL). METHODS We studied 88 randomized APS patients with or without a history of thrombosis. Seventeen patients with other thrombosis (no APS) and 26 healthy subjects served as controls. Platelet adhesion and aggregation on the extracellular matrix were measured with a cone-and-plate(let) analyzer (CPA) by examining the percentage of total area covered with platelets (surface coverage [SC]) and the mean size of surface-bound objects (average size [AS]) and were compared with platelet responses to different ADP concentrations by conventional aggregometry. RESULTS Under defined flow conditions, SC and AS were significantly higher for venous thrombosis and arterial thrombosis in APS patients compared with no thrombosis, other thrombosis, and healthy control groups. The increased platelet adhesion and aggregation in APS patients with thrombotic events was associated with higher levels of von Willebrand factor (vWF) antigen (mean +/- SD 230.6 +/- 51.2%) and ristocetin cofactor activity (181.0 +/- 36.0%). No change in CPA and vWF parameters was found in APS patients with positive results for aPL who did not undergo thrombotic events or in patients with other thrombosis. The CPA parameters were neither associated with the high response of platelets to ADP nor associated with the presence of LAC, aCL, or both. The CPA parameters were similarly increased irrespective of aspirin use. The results suggest that the increased adhesion properties of platelets in APS patients could be mediated by high levels and activity of vWF. This complements the known ability of APS antibodies to enhance platelet response to agonists in conventional aggregometry. CONCLUSION The CPA test was found to be valuable in differentiating APS patients with and without thrombotic complications.
Collapse
Affiliation(s)
- Yair Levy
- Meir Medical Center, Kfar Saba, Israel.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Raymond J, Leblanc P, Chagnon M, Gévry G, Collet JP, Guilbert F, Weill A, Roy D. New Devices Designed to Improve the Long-Term Results of Endovascular Treatment of Intracranial Aneurysms. A Proposition for a Randomized Clinical Trial to Assess their Safety and Efficacy. Interv Neuroradiol 2004; 10:93-102. [PMID: 20587221 DOI: 10.1177/159101990401000201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2004] [Accepted: 03/21/2004] [Indexed: 11/16/2022] Open
Abstract
SUMMARY Endovascular coiling can improve the outcome of patients with ruptured intracranial aneurysms, but angiographic recurrences are frequent compared to surgical clipping. New coils or devices have been introduced to improve long-term results of endovascular treatment but none have been the object of a valid clinical trial. We have proposed a multicentric randomized double-blind study comparing radioactive and standard coil occlusion of aneurysms. The purpose of this article is to review issues that are specific to the design of clinical trials to assess embolic agents that could improve the long-term efficacy of endovascular treatment of intracranial aneurysms. The proposed trial is a randomized, multi-center, prospective, controlled trial comparing the new generation coils to standard platinum coils. Blinding, if at all possible, is preferable to minimize bias, at least for follow-up angiographic studies that should cover a period of 18 months. All patients with an intracranial aneurysm eligible for endovascular treatment would be proposed to participate. The study would enrol approximately 500 patients equally divided between the two groups, recruited within two years, to demonstrate a decrease in the recurrence rate, the primary outcome measure, from 20% to 10%. Secondary outcome measures should assure that complications, initial clinical and angiographic results remain unchanged. Independent data safety and monitoring committees are crucial to the credibility of trials and to ensure scientific rigor and objectivity. The scientific demonstration of an improved long-term efficacy, without significant compromise regarding safety, is mandatory before considering the widespread use of a new embolic device for the endovascular treatment of aneurysms.
Collapse
Affiliation(s)
- J Raymond
- Department of Radiology, Research center of Centre hospitalier de l'Université de Montréal (CHUM) Notre-Dame hospital; Montréal, Quebec, Canada -
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Sakariassen KS, Turitto VT, Baumgartner HR. Recollections of the development of flow devices for studying mechanisms of hemostasis and thrombosis in flowing whole blood. J Thromb Haemost 2004; 2:1681-90. [PMID: 15456474 DOI: 10.1111/j.1538-7836.2004.00907.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
37
|
Thierry B, Merhi Y, Bilodeau L, Trépanier C, Tabrizian M. Nitinol versus stainless steel stents: acute thrombogenicity study in an ex vivo porcine model. Biomaterials 2002; 23:2997-3005. [PMID: 12069342 DOI: 10.1016/s0142-9612(02)00030-3] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Acute and subacute stents thrombosis along with thrombus mediating neointimal proliferation within the stent struts remain major concerns in coronary stenting. Up to date, there is an obvious lack of data on the thrombogenicity of stent materials in physiological conditions. This study was performed to compare the relative thrombogenicity of nitinol versus stainless steel stents. Nitinol stents were laser cut to reproduce the exact geometry of the stainless steel Palmaz stents and tested in an ex vivo AV shunt porcine model under controlled conditions. Nitinol stents presented only small amounts of white and/or red thrombus principally located at the strut intersections while Palmaz stents clearly exhibited more thrombus. As a result, 125I-fibrin(ogen) adsorption and (111)I-platelets adhesion were significantly lower on nitinol than on stainless steel devices (36%, p = 0.03 for fibrin(ogen) and 63%, p = 0.01 for platelet). These results were confirmed by scanning electron observations showing different thrombus morphologies for nitinol and stainless steel. Along with the unique mechanical properties of nitinol, its promising haemocompatibility demonstrated in our study may promote their increasing use for both peripheral and coronary revascularization procedures.
Collapse
Affiliation(s)
- B Thierry
- Department of Biomedical Engineering, McGill University, Montreal , QC, Canada
| | | | | | | | | |
Collapse
|
38
|
Ikarugi H, Shibata M, Ishii K, Yamamoto J. Shear-induced platelet reactivity in middle-aged women after low-intensity exercise. Thromb Res 2001; 104:347-51. [PMID: 11738077 DOI: 10.1016/s0049-3848(01)00375-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- H Ikarugi
- Laboratory of Health and Sport Sciences, Kobe University of Commerce, Kobe, Japan.
| | | | | | | |
Collapse
|
39
|
Sakariassen KS, Hanson SR, Cadroy Y. Methods and models to evaluate shear-dependent and surface reactivity-dependent antithrombotic efficacy. Thromb Res 2001; 104:149-74. [PMID: 11672758 DOI: 10.1016/s0049-3848(01)00344-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The purpose of the present communication is to evaluate the importance of blood flow and surface reactivity for measurement of antithrombotic drug activity or efficacy in selected model systems of thrombus formation. Such information is essential for proper evaluation of antithrombotic drug profiles. The continuous development of flow-dependent thrombosis models for in vitro (anticoagulated blood) and ex vivo (native blood) studies and their application in in vivo animal models from the early 1970s and onwards are briefly considered. Central to this process was the development of various types of perfusion chambers in which a thrombogenic surface is exposed to flowing blood. Such perfusion chambers have been inserted into arteriovenous (AV) shunts in baboon, pig, dog, and rabbit. These approaches have allowed reproducible testing of traditional and novel experimental antithrombotic drugs, and studies on novel drug strategies under well-defined shear conditions and surface reactivity. Shear-dependent antithrombotic efficacy in these models is observed with anticoagulants such as unfractionated heparin, low-molecular weight heparins, or selective inhibitors of thrombin, Factor Xa, or Factor VIIa. However, the degree of shear dependency depends on the nature of the thrombogenic surface, e.g., the inhibition is more pronounced on a tissue factor (TF)-rich surface than on a collagen-rich surface, particularly at venous or low arterial shear. Platelet antagonists such as the COX-1 inhibitor aspirin, inhibitors of thromboxane A2 (TxA2) synthetase, the TxA2 platelet receptor, and of von Willebrand factor (vWf) are shear dependent also, being more efficient at high arterial shear. In contrast, the platelet ADP antagonist clopidogrel, or antagonists to the active platelet membrane glycoprotein IIb-IIIa complex (GPIIb-IIIa) are shear independent. At extremely high arterial shear, which activates platelets and elicit aggregates of circulating platelets, aspirin looses its antithrombotic effect, whereas ADP and GPIIb-IIIa antagonists still interrupt thrombus formation. In general, results obtained with these models mimic and predict antithrombotic efficacy in man when comparison is possible. Information on antithrombotic efficacy in flow devices with various thrombogenic surfaces is now sufficiently available to suggest recommendations for experimental conditions, particularly with regard to blood flow and reactive surfaces.
Collapse
Affiliation(s)
- K S Sakariassen
- Department of Lead Pharmacology, Pharmacia Corporation, Uppsala and Stockholm, Sweden.
| | | | | |
Collapse
|
40
|
Münch K, Wolf MF, Gruffaz P, Ottenwaelter C, Bergan M, Schroeder P, Fogt EJ. Use of simple and complex in vitro models for multiparameter characterization of human blood-material/device interactions. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2001; 11:1147-63. [PMID: 11263805 DOI: 10.1163/156856200744246] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Medical devices, intended for blood contacting applications, undergo extensive in vitro testing followed by animal and clinical feasibility studies. Besides the use of materials known to be intrinsically blood-compatible, the surface of such devices is often modified with a coating in order to improve the performance characteristics during blood exposure. In vitro evaluation of blood-device interactions accompanies the product development cycle from the early design phase using basic material geometries until final finished-product testing. Specific test strategies can vary significantly depending on the end application, the particular study objectives and variables of interest, and cost. To examine the degree to which findings derived from two different in vitro approaches complement one another, this report contrasts findings from a simple multipass loop model with findings from a simulated cardiopulmonary bypass (CPB) model. The loop model consists of tubular test materials, with and without surface modification, formed into valved Chandler loops. The CPB model has an oxygenator with and without surface modification connected to a reservoir and a blood pump. The surface modifications studied in this report are the Carmeda BioActive Surface and Duraflo II heparin coatings. Common blood parameters in the categories of coagulation, platelets, hematology, and immunology were monitored in each model. Ideal models employ the optimal level of complexity to study the design variables of interest and to meet practical cost considerations. In the case of medical device design studies, such models should also be predictive of performance. In the more complex and realistic simulated CPB model, experimental design and cost factors prevented easy/optimum manipulation of critical variables such as blood donor (use of paired samples) and heparin level. Testing in the simpler loop model, on the other hand, readily offered manipulation of these variables, and produced findings which overlapped with observations from the more complex CPB model. Thus, the models described here complimented one another. Moreover, conclusions from consistent findings, such as favorable responses associated with the heparin coatings, between the two models were considered to be more robust.
Collapse
Affiliation(s)
- K Münch
- Materials and Biosciences Center, Medtronic Bakken Research Center, Maastricht, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
41
|
Scrutinio D, Cimminiello C, Marubini E, Pitzalis MV, Di Biase M, Rizzon P. Ticlopidine versus aspirin after myocardial infarction (STAMI) trial. J Am Coll Cardiol 2001; 37:1259-65. [PMID: 11300432 DOI: 10.1016/s0735-1097(01)01164-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVES We sought to compare the efficacy of aspirin and ticlopidine in survivors of acute myocardial infarction (AMI) treated with thrombolysis. BACKGROUND The role of ticlopidine in secondary prevention after AMI has not yet been explored. METHODS Of 4,696 patients with AMI treated with thrombolysis who were screened, 261 died in the hospital (5.6%) and 1,470 were enrolled in this randomized, double-blind, multicenter trial and allocated to treatment with either aspirin (160 mg/day) or ticlopidine (500 mg/day). The most frequent reasons for exclusion were refusal to give informed consent, planned myocardial revascularization, risk of noncompliance with study procedures, need for anticoagulant therapy and contraindications to the study treatments. The primary end point was the first occurrence of any of the following events during the six-month follow-up: fatal and nonfatal AMI, fatal and nonfatal stroke, angina with objective evidence of myocardial ischemia, vascular death or death due to any other cause. RESULTS The primary end point was recorded in 59 (8.0%) of the 736 aspirin-treated and 59 (8.0%) of the 734 ticlopidine-treated patients (p = 0.966). Vascular death was the first event in five patients taking aspirin and in six patients taking ticlopidine (0.7% vs. 0.8%; p = NS); nonfatal AMI in 18 and 8 (2.4% vs. 1.1%; p = 0.049); nonfatal stroke in 3 and 4 (0.4% vs. 0.5%; p = NS); and angina in 33 and 40 (4.5% vs. 5.4%; p = NS), respectively. The frequency of adverse reactions was not significantly different between the two groups. CONCLUSIONS No difference was found between the ticlopidine and aspirin groups in the rate of the primary combined end point of death, recurrent AMI, stroke and angina.
Collapse
Affiliation(s)
- D Scrutinio
- Division of Cardiology, S. Maugeri Foundation, IRCCS, Institute of Rehabilitation, Cassano Murge, Bari, Italy.
| | | | | | | | | | | |
Collapse
|
42
|
Cauwenberghs N, Meiring M, Vauterin S, van Wyk V, Lamprecht S, Roodt JP, Novák L, Harsfalvi J, Deckmyn H, Kotzé HF. Antithrombotic effect of platelet glycoprotein Ib-blocking monoclonal antibody Fab fragments in nonhuman primates. Arterioscler Thromb Vasc Biol 2000; 20:1347-53. [PMID: 10807753 DOI: 10.1161/01.atv.20.5.1347] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Platelet adhesion in arterial blood flow is mainly supported by the platelet receptor glycoprotein (GP) Ib, which interacts with von Willebrand factor (vWF) that is bound to collagen at the site of vessel wall injury. Antibody 6B4 is a monoclonal antibody (MoAb) raised against purified human GPIb. MoAb 6B4 inhibits both ristocetin- and botrocetin-induced, vWF-dependent human platelet agglutination. MoAb 6B4 furthermore blocks shear-induced adhesion of human platelets to collagen I. We studied the antithrombotic effect of this inhibitory murine MoAb 6B4 in a baboon model of arterial thrombosis. When injected into baboons, intact IgG and its F(ab')(2) fragments caused almost immediate thrombocytopenia, whereas injection of the Fab fragments alone did not. Fab fragments were subsequently used to investigate their in vivo effect on platelet deposition onto a thrombogenic device, consisting of collagen-rich, glutaraldehyde-fixed bovine pericardium (0.6 cm(2)), at a wall shear rate ranging from 700 to 1000 s(-1). Baboons were either pretreated with Fabs to study the effect of inhibition on platelet adhesion or treated 6 minutes after placement of the thrombogenic device to investigate the effect on interplatelet cohesion. Pretreatment of the animals with bolus doses ranging from 80 to 640 microgram/kg Fab fragments significantly reduced (111)In-labeled platelet deposition onto the collagen surface by approximately 43% to 65%. Only the highest dose caused a significant prolongation (doubling) of the bleeding time. Ex vivo ristocetin-induced platelet agglutination was equally reduced. Treatment with a bolus of 110 microgram/kg Fab fragments after a thrombus was allowed to form for 6 minutes had no effect on further platelet deposition. We therefore conclude that Fab fragments or derivatives of inhibitory anti-GPIb antibodies may be useful compounds to prevent thrombosis.
Collapse
Affiliation(s)
- N Cauwenberghs
- Laboratory for Thrombosis Research, Interdisciplinary Research Center, KU Leuven Campus Kortrijk, Kortrijk, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Bossavy JP, Sakariassen KS, Thalamas C, Boneu B, Cadroy Y. Antithrombotic efficacy of the vitamin K antagonist fluindione in a human Ex vivo model of arterial thrombosis : effect of anticoagulation level and combination therapy with aspirin. Arterioscler Thromb Vasc Biol 1999; 19:2269-75. [PMID: 10479672 DOI: 10.1161/01.atv.19.9.2269] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Thrombin is a main mediator of arterial thrombus formation, and its inhibition is an important antithrombotic strategy. However, the place of vitamin K antagonists among the different therapeutic strategies for preventing arterial thrombus formation is still debated. We studied the antithrombotic efficacy of the vitamin K antagonist fluindione in a human ex vivo model of arterial thrombosis and determined whether aspirin enhances fluindione efficacy. Ten healthy male volunteers were randomly assigned to receive fluindione, alone or in combination with aspirin (325 mg/d). Fluindione was given at increasing doses to give a stable international normalized ratio (INR) between 1.5 and 2.0 and between 2.1 and 3.0. We induced arterial thrombus formation ex vivo by exposing collagen- or tissue factor (TF)-coated coverslips in a parallel-plate perfusion chamber to native blood for 3 minutes at an arterial wall shear rate of 2600 s(-1). Platelet and fibrin deposition were measured by immunoenzymatic methods. Fluindione inhibited thrombus formation on TF-coated coverslips in a dose-dependent manner by 50% and 80% at INR 1.5 to 2.0 and INR 2.1 to 3.0, respectively (P<0.05). Fluindione in combination with aspirin inhibited TF-induced thrombus formation in a comparable manner. Collagen-induced thrombus formation was not reduced in subjects treated by fluindione. It was reduced by 50% to 60% in those treated with fluindione plus aspirin, regardless of the level of anticoagulation (P<0.05). Thus, the effectiveness of fluindione for preventing arterial thrombosis is dependent on the nature of the thrombogenic trigger. Fluindione is very effective in preventing TF- but not collagen-triggered thrombus formation. Aspirin enhances the antithrombotic effectiveness of fluindione, because combined treatment interrupts both TF- and collagen-induced thrombus formation.
Collapse
Affiliation(s)
- J P Bossavy
- Laboratoire de Recherche sur l'Hémostase et la Thrombose, Pavillon Lefèbvre, CHU Purpan, Toulouse France
| | | | | | | | | |
Collapse
|
44
|
Bossavy JP, Sakariassen KS, Rübsamen K, Thalamas C, Boneu B, Cadroy Y. Comparison of the antithrombotic effect of PEG-hirudin and heparin in a human ex vivo model of arterial thrombosis. Arterioscler Thromb Vasc Biol 1999; 19:1348-53. [PMID: 10323789 DOI: 10.1161/01.atv.19.5.1348] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Polyethylene glycol (PEG)-hirudin is a derivative of hirudin with a long plasma half-life. We have compared the efficacy of PEG-hirudin with unfractionated heparin (UH) in preventing arterial thrombosis. Arterial thrombus formation was induced ex vivo in 12 healthy human volunteers by exposing a tissue factor-coated coverslip positioned in a parallel-plate perfusion chamber to flowing nonanticoagulated human blood drawn directly from an antecubital vein at an arterial wall shear rate of 2600 s-1 for 3.5 minutes. PEG-hirudin, UH, or saline (as control) were administered ex vivo through a heparin-coated mixing device positioned proximal to the perfusion chamber. Platelet and fibrin deposition was quantified by immunoenzymatic measure of the P-selectin and D-dimer content of dissolved plasmin-digested thrombi, respectively. UH was administered to a plasma concentration of 0.35 IU/mL. This concentration prolonged the activated partial thromboplastin time from 32+/-1 seconds to 79+/-4 seconds (P<0.01). UH did not significantly prevent platelet deposition. However, fibrin deposition was reduced by 39% (P<0.05). PEG-hirudin in plasma concentrations of 0.5, 2.5, and 5 microg/mL prolonged the activated partial thromboplastin time to 48+/-2, 87+/-4, and 118+/-4 seconds, respectively. In contrast to UH, PEG-hirudin prevented both platelet and fibrin deposition in a dose-dependent manner with a >80% reduction at 5 microg/mL (P<0.01). Furthermore, the plasma level of PEG-hirudin required to significantly prevent fibrin deposition (0.5 microg/mL) corresponded to a much shorter prolongation of activated partial thromboplastin time (48+/-2 seconds) than that needed for UH (79+/-4 seconds). Thus, our results are compatible with the view that thrombin is greatly involved in recruitment of platelets in evolving thrombi, and that PEG-hirudin is an effective agent for preventing arterial thrombosis in a human ex vivo experimental model.
Collapse
Affiliation(s)
- J P Bossavy
- Service de Chirurgie Générale et Vasculaire, Toulouse, France
| | | | | | | | | | | |
Collapse
|
45
|
Anderson JM, Bianco RW, Grehan JF, Grubbs BC, Hanson SR, Hauch KD, Lahti M, Mrachek JP, Northup SJ, Ratner BD, Schoen FJ, Schroeder EL, Schumacher CW, Svendsen CA. Biological Testing of Biomaterials. Biomater Sci 1996. [DOI: 10.1016/b978-012582460-6/50008-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|