Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Eur Heart J ; 23(15): 1202-12, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12127922

ABSTRACT

AIMS: We compared the effects of hirudin and heparin on thrombin generation and activity among 350 patients with acute coronary syndromes enrolled in the GUSTO-IIb trial. METHODS AND RESULTS: We obtained blood at baseline; at 4, 8, and 24h into infusion; at drug termination; and 6 and 24h after termination. We assayed for thrombin activity (fibrinopeptide A, activated protein C, thrombin-antithrombin complex), thrombin generation (prothrombin fragment 1.2), and platelet activation (platelet factor 4). Median baseline fibrinopeptide A and platelet factor 4 levels were elevated. Thrombin formation tended to increase with hirudin and decrease with heparin; by 8h into infusion, thrombin formation was significantly less for heparin (P<0.01). Most patients showed reduced thrombin activity and platelet activation during infusion of either agent. Hirudin-assigned patients had significantly lower fibrinopeptide A levels during infusion. Six h post-termination, both groups had increased thrombin activity. Thrombin formation was increased in heparin patients (P<0.0001), significantly more than with hirudin (P=0.005). Higher values of haemostasis markers tended to be associated with poorer 30-day outcomes. CONCLUSION: Although hirudin did not prevent generation of new thrombin, it appeared to inhibit thrombin activity more than did heparin and produced slower increases in thrombin formation after discontinuation. The reelevation of thrombotic markers after stopping intravenous antithrombin therapy and the tendency toward increased thrombotic events with post-treatment increases in marker levels suggest an ongoing, clinically significant prothrombotic state. These results raise the possibility of improving on current antithrombotics by preventing thrombin generation and thrombin activity and by sustained suppression of the prothrombotic state.


Subject(s)
Fibrinolytic Agents/pharmacology , Hemostasis/drug effects , Heparin/pharmacology , Hirudins/pharmacology , Acute Disease , Aged , Blood Coagulation/drug effects , Double-Blind Method , Female , Fibrinolytic Agents/therapeutic use , Heart Diseases/blood , Heart Diseases/drug therapy , Heparin/therapeutic use , Hirudin Therapy , Humans , Male , Middle Aged , Syndrome , Thrombin/metabolism , Treatment Outcome
2.
Proc Natl Acad Sci U S A ; 98(7): 4078-83, 2001 Mar 27.
Article in English | MEDLINE | ID: mdl-11274432

ABSTRACT

Tissue factor (TF), the initiator of blood coagulation and thrombosis, is up-regulated after vascular injury and in atherosclerotic states. Systemic administration of recombinant TF pathway inhibitor (TFPI) has been reported to decrease intimal hyperplasia after vascular injury and also to suppress systemic mechanisms of blood coagulation and thrombosis. Here we report that, in heritable hyperlipidemic Watanabe rabbits, adenoviral gene transfer of TFPI to balloon-injured atherosclerotic arteries reduced the extent of intimal hyperplasia by 43% (P < 0.05) compared with a control vector used at identical titer (1 x 10(10) plaque-forming units/ml). Platelet aggregation and coagulation studies performed 7 days after local gene transfer of TFPI failed to show any impairment in systemic hemostasis. At time of sacrifice, 4 weeks after vascular injury, the 10 Ad-TFPI treated carotid arteries were free of thrombi, whereas two control-treated arteries were occluded (P, not significant). These findings suggest that TFPI overexpressed in atherosclerotic arteries can regulate hyperplastic response to injury in the absence of changes in the hemostatic system, establishing a role for local TF regulation as target for gene transfer-based antirestenosis therapies.


Subject(s)
Arteriosclerosis/prevention & control , Lipoproteins/therapeutic use , Tunica Intima/pathology , Adenoviridae/genetics , Angioplasty, Balloon, Coronary/adverse effects , Animals , Arteriosclerosis/etiology , Arteriosclerosis/pathology , Disease Models, Animal , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Humans , Hyperplasia/prevention & control , Lipoproteins/genetics , Lipoproteins/metabolism , Muscle, Smooth, Vascular/metabolism , Rabbits
3.
Circulation ; 103(3): 407-14, 2001 Jan 23.
Article in English | MEDLINE | ID: mdl-11157693

ABSTRACT

BACKGROUND: The transcription factor E2F-1 promotes S-phase entry and death in transformed cells and primary cardiomyocytes. We tested the hypothesis that overexpression of E2F-1 forces growth-arrested human coronary vascular smooth muscle cells (VSMCs) to enter the S phase, undergo apoptosis, and thereby regulate VSMC growth. METHODS AND RESULTS: Early-passage (8 times. CONCLUSIONS: Overexpression of the transcription factor E2F-1 regulates growth of human coronary VSMCs by forcing the cells to enter the S phase and then to die. Cell death appears to involve caspase 3-like activity, which, in the VSMCs, is markedly increased by overexpression of E2F-1.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Coronary Vessels/growth & development , DNA-Binding Proteins , Muscle Development , Muscle, Smooth, Vascular/growth & development , Transcription Factors/genetics , Adenoviridae/genetics , Apoptosis , Caspase 3 , Caspases/biosynthesis , Cell Count , Cells, Cultured , Coronary Vessels/cytology , E2F Transcription Factors , E2F1 Transcription Factor , Enzyme Induction , Flow Cytometry , Genetic Vectors , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Muscle, Smooth, Vascular/cytology , Retinoblastoma-Binding Protein 1 , S Phase , Time Factors , Transcription Factor DP1 , Transcription Factors/biosynthesis , Transcription Factors/pharmacology , Transfection
4.
Am J Physiol Heart Circ Physiol ; 279(6): H3065-75, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11087265

ABSTRACT

We tested the hypothesis that selectin inhibition with blocking antibodies or a small-molecular-weight inhibitor of L-, P-, and E-selectin, methoxybenzoylpropionic acid (MBPA), prevents thrombus formation in a canine coronary Folts' model. Cyclic flow variations (CFVs) were induced by crush injury and constriction of the left anterior descending coronary artery in dogs. Systemic infusion of antibodies to P- and L-selectin abolished CFVs, respectively, in 50% and 17% of treated dogs [P = not significant (NS)]. The combination of P- and L-selectin antibodies suppressed CFVs in 60% of treated dogs (P = NS). In contrast, systemic selectin blockade by intravenous infusion or local adventitial application of MBPA markedly reduced CFVs and, in addition, reduced myocardial myeloperoxidase (MPO) activity. We conclude that inhibition of L-, P-, and E-selectin binding by a small-molecular-weight, noncarbohydrate compound markedly reduces arterial thrombosis, whereas systemic administration of antibodies to L- and P-selectin fail to reproduce this antithrombotic effect. These results underscore the role of selectins in the pathogenesis of arterial thrombosis under high shear stress and suggest that inhibition of P- and L- selectin may not suffice to prevent thrombus formation in this model. The role of E-selectin in thrombus formation in this model awaits further testing.


Subject(s)
Coronary Thrombosis/drug therapy , Coronary Thrombosis/immunology , Propionates/pharmacology , Selectins/immunology , Animals , Antibodies, Monoclonal/pharmacology , Blood Cell Count , Coronary Thrombosis/prevention & control , Coronary Vessels/injuries , Coronary Vessels/physiopathology , Disease Models, Animal , Dogs , E-Selectin/immunology , Female , Injections, Intravenous , L-Selectin/immunology , Leukocytes/immunology , Male , Neutralization Tests , Neutrophils/immunology , P-Selectin/immunology , Phenyl Ethers , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Vasculitis/drug therapy , Vasculitis/immunology , Vasculitis/prevention & control
5.
Circulation ; 101(3): 289-95, 2000 Jan 25.
Article in English | MEDLINE | ID: mdl-10645925

ABSTRACT

BACKGROUND: Tissue factor pathway inhibitor (TFPI) is an endogenous inhibitor of factor Xa and the coagulant initiator complex tissue factor/factor VIIa. METHODS AND RESULTS: To study the effects of TFPI gene transfer on thrombus formation, balloon-injured porcine carotid arteries were treated locally with an adenovirus encoding human TFPI (Ad-TFPI) or control virus. Gene transfer of TFPI was confirmed by detection of human TFPI in the conditioned medium of porcine carotid arteries kept in culture after in vivo transduction. When carotid flow was measured with Doppler probe 5 days after surgery, cyclic flow variations (CFVs) developed in 7 of 8 control pigs after constriction of the injured carotid artery by 40%, and all control-treated arteries occluded after 70% constriction. In contrast, CFVs were observed in only 1 of 8 Ad-TFPI-treated pigs after 40% constriction, and only 3 of 8 occluded after constriction by 70% (P=0.0027 and P=0.007, respectively). None of the 5 TFPI-transduced arteries open after 70% constriction developed CFVs during an incremental epinephrine infusion. CONCLUSIONS: Compared with baseline, systemic hemostatic variables and platelet aggregation were unimpaired, suggesting that TFPI gene transfer can prevent arterial thrombosis in the presence of severe shear stress and without detectable hemostatic impairment.


Subject(s)
Carotid Artery Diseases/prevention & control , Factor Xa Inhibitors , Genetic Therapy , Lipoproteins/genetics , Thrombosis/prevention & control , Adenoviridae/genetics , Animals , Catheterization , Cerebrovascular Circulation , Humans , Lipoproteins/physiology , Male , Swine
6.
Z Kardiol ; 89(12): 1072-83, 2000 Dec.
Article in German | MEDLINE | ID: mdl-11201022

ABSTRACT

Despite considerable progress, pharmacological therapies have not provided a complete solution for common cardiovascular problems, including recurrent thrombosis, restenosis, and vein graft deterioration. Optimal drug dosage, reproducing plasma concentrations achieved in animal studies establishing proof-of-principle, would often be too toxic to administer. Local gene therapy aims at overexpressing proteins that regulate the cell cycle of vascular smooth muscle cells, inhibit vascular smooth muscle cell migration, endow the endothelium with enhanced vasoprotective properties. Alternatively, some approaches tend to suppress gene expression of proteins believed to promote vascular smooth muscle cell proliferation and migration. In sharp contrast to drug treatments, local gene therapy limits expression of the beneficial agent to the injured vascular site, where it can extend the presence of this agent to weeks and, with some gene vectors, to many months. This review summarizes and discusses antithrombotic gene therapy approaches for the prevention of restenosis and late thrombosis after catheter-based revascularizations.


Subject(s)
Coronary Disease/therapy , Coronary Thrombosis/therapy , Genetic Therapy , Angioplasty, Balloon, Coronary , Animals , Coronary Disease/genetics , Coronary Thrombosis/genetics , Humans , Recurrence
7.
Semin Interv Cardiol ; 4(3): 151-65, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10545621

ABSTRACT

Despite considerable progress, pharmacological therapies have not provided a complete solution for common cardiovascular problems, including recurrent thrombosis, restenosis, and vein graft deterioration. Optimal drug dosage, reproducing plasma concentrations achieved in animal studies establishing proof-of-principle, would often be too toxic to administer, especially when given over prolonged periods of time. Local gene therapy aims at overexpressing proteins that: (1) regulate the cell cycle of VSMC; (2) inhibit VSMC migration; (3) endow the endothelium with its vasoprotective properties; and (4) stimulate growth of endothelium and angiogenesis. Alternatively, some approaches tend to suppress gene expression of proteins believed to promote VSMC proliferation and migration. In sharp contrast to drug treatments, local gene therapy limits expression of the beneficial agent to the injured vascular site, and there, it can extend the presence of this agent to weeks and, with some gene vectors, to many months. The clinical potential of this approach has led to the initiation of trials that currently evaluate gene therapy approaches to the attenuation of peripheral and myocardial ischaemia and the prevention of vein graft disease.


Subject(s)
Coronary Disease/therapy , Genetic Therapy/methods , Animals , Clinical Trials as Topic , Disease Models, Animal , Female , Genetic Therapy/adverse effects , Humans , Male , Prognosis , Treatment Outcome
8.
Clin Cardiol ; 22(8 Suppl): IV44-53, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10492853

ABSTRACT

An extensive body of research conducted in the past 25 years has helped foster understanding of the mechanisms and pathogenesis of the acute coronary syndromes and occlusive disease. Thus, it is well established that thrombosis is caused by vascular injury and that immediate lysis of the arterial thrombus and subsequent prevention of thrombotic reocclusion are critical to the treatment of these disorders. Remarkable progress in the understanding of the biological and molecular mechanisms involved in vascular-wall-platelet interactions, platelet-platelet interactions, and coagulation has led to the identification of multiple targets for drug discovery and the development of numerous antithrombotic drugs. The purpose of this article is to review emerging antithrombotic therapies, introduce potential future molecular targets for drug discovery efforts, and discuss novel strategies for managing patients with coronary disease.


Subject(s)
Coronary Disease/drug therapy , Coronary Thrombosis/etiology , Fibrinolytic Agents/therapeutic use , Thrombolytic Therapy/trends , Clopidogrel , Coronary Thrombosis/physiopathology , Cyclooxygenase 2 , Enzyme Inhibitors/therapeutic use , Forecasting , Humans , Isoenzymes , Membrane Proteins , P-Selectin , Platelet Activation , Platelet Aggregation Inhibitors/therapeutic use , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Prostaglandin-Endoperoxide Synthases , Ticlopidine/analogs & derivatives , Ticlopidine/therapeutic use
9.
Circulation ; 98(9): 919-26, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9738648

ABSTRACT

BACKGROUND: Nitric oxide, synthesized from L-arginine by nitric oxide synthase (NOS), is a vasodilator and inhibits vascular smooth muscle cell (SMC) proliferation and migration. The effects of local NOS gene transfer on restenosis after experimental balloon angioplasty were investigated. METHODS AND RESULTS: Left anterior descending coronary artery angioplasty was performed in 25 pigs. Animals received an intramural injection of adenovirus (1.5 x 10(9) pfu) carrying either the NOS cDNA (AdCMVceNOS) or no cDNA (AdRR5) via the Infiltrator. Local gene transfer efficiency and bioactivity of recombinant protein were assessed after 4 days. Indices of restenosis were evaluated by computerized planimetry on coronary artery sections prepared 28 days after angioplasty. Adenoviral vectors permitted efficient gene delivery to medial SMCs and adventitial cells of coronary arteries. Vascular cGMP levels were depressed after angioplasty from 1.30+/-0.42 to 0.33+/-0.20 pmol/mg protein (P<0.05) but were restored after constitutive endothelial (ce) NOS gene transfer to 1.82+/-0.98 pmol/mg (P<0.05 versus injured group and P=NS versus control). The ratio of the neointimal area to the internal elastic lamina fracture length, maximal neointimal thickness, and percent stenosis were all reduced in AdCMVceNOS- versus AdRR5-transduced pigs (0.59+/-0.14 versus 0.80+/-0.19 mm, P=0.02; 0.75+/-0.21 versus 1.04+/-0.25 mm, P=0.019; and 53+/-15% versus 75+/-11%, P=0.006, respectively). Lumen area was significantly larger (0.70+/-0.35 mm2 in AdCMVceNOS versus 0.32+/-0.18 mm2 in AdRR5, P=0.007). CONCLUSIONS: Percutaneous adenovirus-mediated NOS gene transfer resulted in efficient local overexpression of functional NOS after angioplasty in coronary arteries. Restored NO production in injured coronary arteries significantly reduced luminal narrowing, most likely through a combined effect on neointima formation and on vessel remodeling after angioplasty.


Subject(s)
Adenoviridae , Angioplasty, Balloon, Coronary/adverse effects , Coronary Vessels/injuries , Gene Transfer Techniques , Nitric Oxide Synthase/genetics , Animals , Coronary Vessels/enzymology , Coronary Vessels/physiopathology , Cyclic GMP/metabolism , Gene Expression , Genes, Reporter , Muscle, Smooth, Vascular/enzymology , Myocardial Ischemia/therapy , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III , Recurrence , Swine , Transgenes/genetics , Tunica Intima/enzymology , beta-Galactosidase/genetics
10.
Circulation ; 97(13): 1274-81, 1998 Apr 07.
Article in English | MEDLINE | ID: mdl-9570198

ABSTRACT

BACKGROUND: Loss of endothelial NO production after arterial injury may contribute to restenosis, characterized by neointima formation and elastic recoil. Adenovirus-mediated transfer of the gene encoding NO synthase (NOS) in balloon-injured arteries may restore NO production and inhibit neointima formation. METHODS AND RESULTS: After balloon injury, rat carotid arteries were transduced with 3x10(10) pfu/mL recombinant adenovirus carrying the human endothelial constitutive NOS cDNA (AdCMVceNOS, n=8) or no cDNA (AdRR5, n=8). ceNOS expression was confirmed by immunoblot analysis of vascular extracts and was localized by immunostaining in 30% of medial smooth muscle cells (SMCs) and in the adventitia of AdCMVceNOS-transduced arteries. Vascular cGMP levels were reduced from 3.9 pmol/g wet wt in uninjured arteries to 0.7 pmol cGMP/g after AdRR5 but were restored after ceNOS gene transfer (3.8 pmol cGMP/g wet wt, P<.05 versus AdRR5). Intima-to-media ratio 2 weeks after injury was significantly reduced (0.19+/-0.02 in AdCMVceNOS-infected versus 0.69+/-0.07 in AdRR5-infected arteries, P<.05). In vitro, BrdU incorporation of AdCMVceNOS-infected SMCs was reduced by 28% compared with AdRR5-infected SMCs. Transduced cells from injured carotid arteries subjected to FACS sorting showed a significantly lower BrdU labeling index in ceNOS-infected rats (29+/-6% versus 43+/-5% and 45+/-4% in control, injured, and AdRR5-infected rats, respectively, P<.05). CONCLUSIONS: AdCMVceNOS gene transfer to balloon-injured rat carotid arteries restores vascular NO production and reduces neointima formation, at least in part because of an antiproliferative effect on medial SMCs. Adenovirus-mediated ceNOS gene transfer might reduce arterial restenosis after balloon angioplasty.


Subject(s)
Angioplasty, Balloon/adverse effects , Endothelium, Vascular/enzymology , Gene Transfer Techniques , Muscle, Smooth, Vascular/cytology , Nitric Oxide Synthase/genetics , Tunica Intima/injuries , Adenoviridae/genetics , Animals , Aorta/injuries , Aorta/pathology , Cell Division/physiology , Humans , Nitric Oxide Synthase Type III , Rats , Tunica Intima/pathology
11.
J Clin Invest ; 98(2): 317-24, 1996 Jul 15.
Article in English | MEDLINE | ID: mdl-8755640

ABSTRACT

Nitric oxide (NO), a vasodilator involved in the regulation of pulmonary vascular tone, is synthesized by a family of enzymes, nitric oxide synthases (NOS). To investigate whether adenoviral-mediated overexpression of constitutive endothelial NOS (ceNOS) would attenuate hypoxic pulmonary vasoconstriction, we aerosolized 3 X 10(9) plaque forming units of a recombinant adenovirus containing the ceNOS gene (AdCMVceNOS) into rat lungs. Four days after infection, transgene expression was confirmed using immunoblot techniques. Diffuse ceNOS immunostaining was detected in alveoli and medium-sized and small pulmonary vessels of AdCMVceNOS-transduced lungs. AdCMVceNOS-transduction was associated with an 86% increase in [3H]arginine to [3H]citrulline conversion and a rise in pulmonary cGMP levels from 7 +/- 1 to 59 +/- 9 pmol/mg protein in lungs from AdCMVceNOS versus control rats, (P < 0.05). During acute hypoxia (FIO2 = 0.10) for 25 min, mean pulmonary artery pressure (PAP) increased significantly from 17 +/- 1 to 27 +/- 1 mmHg in rats aerosolized with saline (n = 4) and from 18 +/- 1 to 28 +/- 1 mmHg in rats given an adenoviral vector expressing a nuclear-targeted beta-galactosidase gene (AdCMV beta gal, n = 8). In contrast, in AdCMVceNOS-transduced rats (n = 8) the hypoxia-induced increase in PAP was significantly attenuated (18 +/- 1 to 23 +/- 2 mmHg). Systemic blood pressure was not affected by aerosol gene transfer. Thus, adenoviral-mediated ceNOS gene transfer to rat lungs increases ceNOS expression and activity, and reduces acute hypoxic pulmonary vasoconstriction. Aerosolized recombinant adenovirus overexpressing vasodilatory proteins can act as a selective pulmonary vasodilator and may hold promise as a future therapeutic strategy for pulmonary hypertension.


Subject(s)
Endothelium, Vascular/enzymology , Gene Transfer Techniques , Lung/physiology , Nitric Oxide Synthase/biosynthesis , Pulmonary Circulation/physiology , Vasoconstriction , Adenoviridae , Analysis of Variance , Animals , Base Sequence , Cell Line , Cytomegalovirus , DNA Primers , Genetic Vectors , Humans , Hypoxia , Immunohistochemistry , Kinetics , Lung/blood supply , Molecular Sequence Data , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase/genetics , Polymerase Chain Reaction , Rats , Rats, Wistar , Restriction Mapping
12.
Circulation ; 93(4): 792-9, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8641009

ABSTRACT

BACKGROUND: Thrombolysis to normal flow in patients with acute myocardial infarction preserves left ventricular function and decreases mortality. Failure of early reperfusion, reocclusion, or residual thrombus may be due to concurrent activation of the platelet-coagulation system. Thus, we hypothesized that the best concomitant antithrombotic therapy (recombinant [r]-hirudin, heparin, or aspirin) will maximally accelerate thrombolysis by r-tissue-type plasminogen activator (rTPA) and reduce residual thrombus. METHODS AND RESULTS: Occlusive thrombi were formed in the carotid arteries of 29 pigs (by balloon dilatation followed by endarterectomy at the site of injury-induced vasospasm) and matured for 30 minutes before rTPA was started, with or without antithrombotic therapy. Thrombolysis was assessed with the use of angiography and measurement of residual thrombus. Pigs were allocated to one of five treatments: placebo, rTPA, rTPA plus r-hirudin, rTPA plus heparin, or rTPA plus intravenous aspirin. No placebo-treated pig reperfused. Two of six animals treated with rTPA alone reperfused compared with seven of seven animals treated with rTPA plus r-hirudin (reperfusion time, 33 +/- 10 minutes), six of seven animals treated with rTPA plus heparin (reperfusion time, 110 +/- 31 minutes), and two of six animals with rTPA plus aspirin. The activated partial thromboplastin time was prolonged in only the rTPA plus r-hirudin group (25 +/- 0.1 times baseline) and the rTPA plus heparin group (5.3 +/- 0.2 times baseline). Residual 111In-platelet and 125I-fibrin(ogen) depositions were lower in the heparin-treated group and lowest in the r-hirudin-treated group (heparin versus hirudin, respectively; incidence of residual macroscopic thrombus was six of six animals versus two of seven [P = .01]; 125I-fibrin(ogen), 170 +/- 76 versus 48 +/- 6 x 10(6) molecules/cm2 [P = .02]; 111In-platelets, 47 +/- 15 versus 13 +/- 2 x 10(6)/cm2, P = .10). No pigs developed spontaneous bleeding. CONCLUSIONS: Thrombin inhibition with heparin or r-hirudin significantly accelerated thrombolysis of occlusive platelet-rich thrombosis, but only the best antithrombotic therapy (r-hirudin) eliminated or nearly eliminated residual thrombus.


Subject(s)
Aspirin/therapeutic use , Fibrinolytic Agents/therapeutic use , Heparin/therapeutic use , Hirudin Therapy , Thrombosis/drug therapy , Animals , Aspirin/administration & dosage , Blood Platelets/drug effects , Blood Platelets/pathology , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Carotid Arteries/pathology , Drug Therapy, Combination , Fibrin/metabolism , Fibrinolytic Agents/administration & dosage , Hemostasis , Heparin/administration & dosage , Hirudins/administration & dosage , Humans , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Swine , Thrombosis/pathology , Time Factors , Tissue Plasminogen Activator/administration & dosage , Tissue Plasminogen Activator/therapeutic use
13.
Circulation ; 93(1): 10-7, 1996 Jan 01.
Article in English | MEDLINE | ID: mdl-8616915

ABSTRACT

BACKGROUND: Prostacyclin is an important vasoprotective molecule. It inhibits platelet aggregation, monocyte interaction with endothelium, and smooth muscle cell lipid accumulation. Vascular cyclooxygenase-1 (COX-1) is the rate-limiting step in prostacyclin synthesis. The objective of this study was to determine whether adenovirus-mediated transfer of COX-1 could restore COX-1 activity, augment prostacyclin synthesis, and prevent thrombus formation in a porcine carotid angioplasty model. METHODS AND RESULTS: Human COX-1 cDNA driven by a cytomegalovirus promoter was constructed into a replication-defective adenovirus 5 vector by homologous recombination. Recombinant adenovirus without a foreign gene (Ad-RR) and buffer were included as controls. Recombinant Ad-LacZ was used for marking the transfected cells in vivo. In the in vitro experiments, cultured human endothelial cells (ECs) and porcine arterial smooth muscle cells (SMCs) were incubated with Ad-COX-1 for 2 hours and 6-keto-PGF(1 alpha) level and the transgene expression were determined 72 hours after infection. In the in vivo experiments, recombinant adenoviruses were directly instilled into angioplasty-injured porcine carotid arteries for 30 minutes. Cyclic flow changes were monitored for 10 days and thrombus formation was examined histologically thereafter. Transgene expression and prostaglandin I2 (PGI2) synthesis by the injured arteries were determined. Cultured ECs infected with Ad-COX-1 produced a fivefold to eightfold increase in PGI2, and the transgene expression in cultured porcine SMCs was demonstrated by Northern analysis. Direct administration of Ad-COX-1 at a dose of 3 x 10(10) pfu completely inhibited carotid cyclic flow changes and thrombus formation accompanied by a fourfold increase in PGI2 synthesis by the injured arteries 10 days after infection, whereas Ad-COX-1 at a lower dose, 5 x 10(9) pfu, had no antithrombotic effects when compared with Ad-RR vector and buffer controls. CONCLUSIONS: Adenovirus-mediated transfer of COX-1 to angioplasty-injured carotid arteries was efficacious in augmenting PGI2 synthesis and was associated with an inhibition of thrombosis when a relatively high titer of adenovirus was instilled.


Subject(s)
Carotid Artery Thrombosis/prevention & control , Epoprostenol/biosynthesis , Genetic Therapy , Muscle, Smooth, Vascular/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Adenoviridae/genetics , Angioplasty, Balloon/adverse effects , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Thrombosis/etiology , Carotid Artery Thrombosis/metabolism , Epoprostenol/therapeutic use , Gene Transfer Techniques , Genetic Vectors , Humans , Muscle, Smooth, Vascular/pathology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Recombinant Proteins/biosynthesis , Swine
14.
Drugs ; 49(6): 856-84, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7641602

ABSTRACT

Platelet activation plays a critical role in thromboembolic disorders, and aspirin remains a keystone in preventive strategies. This remarkable efficacy is rather unexpected, as aspirin selectively inhibits platelet aggregation mediated through activation of the arachidonic-thromboxane pathway, but not platelet aggregation induced by adenosine diphosphate (ADP), collagen and low levels of thrombin. This apparent paradox has stimulated investigations on the effect of aspirin on eicosanoid-independent effects of aspirin on cellular signalling. It has also fostered the search for antiplatelet drugs inhibiting platelet aggregation at other levels than the acetylation of platelet cyclo-oxygenase, such as thromboxane synthase inhibitors and thromboxane receptor antagonists. The final step of all platelet agonists is the functional expression of glycoprotein (GP) IIb/IIIa on the platelet surface, which ligates fibrinogen to link platelets together as part of the aggregation process. Agents that interact between GPIIb/IIIa and fibrinogen have been developed, which block GPIIb/IIIa, such as monoclonal antibodies to GPIIb/IIIa, and natural and synthetic peptides (disintegrins) containing the Arg-Gly-Asp (RGD) recognition sequence in fibrinogen and other adhesion macromolecules. Also, some non-peptide RGD mimetics have been developed which are orally active prodrugs. Stable analogues of prostacyclin, some of which are orally active, are also available. Thrombin has a pivotal role in both platelet activation and fibrin generation. In addition to natural and recombinant human antithrombin III, direct antithrombin III-independent thrombin inhibitors have been developed as recombinant hirudin, hirulog, argatroban, boroarginine derivatives and single stranded DNA oligonucleotides (aptanes). Direct thrombin inhibitors do not affect thrombin generation and may leave some 'escaping' thrombin molecules unaffected. Inhibition of factor Xa can prevent thrombin generation and disrupt the thrombin feedback loop that amplifies thrombin production.


Subject(s)
Fibrinolytic Agents/pharmacology , Amino Acid Sequence , Animals , Clinical Trials as Topic , Clopidogrel , Fibrinolytic Agents/therapeutic use , Glycoproteins/metabolism , Molecular Sequence Data , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Protein C/metabolism , Thrombomodulin , Thrombosis/drug therapy , Thrombosis/prevention & control , Ticlopidine/analogs & derivatives , Ticlopidine/pharmacology
16.
Trans Am Clin Climatol Assoc ; 106: 100-7; discussion 107-8, 1995.
Article in English | MEDLINE | ID: mdl-7483165

ABSTRACT

These preliminary studies demonstrate the feasibility of restoration of prostacyclin synthesis in mechanically-injured porcine carotid arteries following angioplasty. Our initial data suggest the possibility of inhibiting thrombus development by adenovirus-CMV-PGHS-1 therapy in the initial 10 days following angioplasty.


Subject(s)
Endothelium, Vascular/injuries , Endothelium, Vascular/metabolism , Epoprostenol/metabolism , Genetic Therapy/methods , Adenoviridae/genetics , Animals , Carotid Arteries/metabolism , Carotid Artery Injuries , Coronary Disease/metabolism , Coronary Disease/therapy , Humans , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Swine
17.
Circulation ; 90(6): 2671-8, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7994807

ABSTRACT

BACKGROUND: The degree to which antithrombotic drugs suppress thrombin generation is unknown. Because hirudin, unlike antithrombin III, binds intravascular thrombin rapidly and selectively to yield a circulating inactive complex of 3- to 4-hour half-life, we used intravenous hirudin in humans to investigate the course of thrombin generation during and early after anticoagulation with this potent, direct antithrombin. METHODS AND RESULTS: Intravascular thrombin was measured with an ELISA for the thrombin-hirudin complex formed during and for 18 hours after stopping a 6-hour infusion of hirudin at 0.1, 0.2, and 0.3 mg.kg-1.h-1 in three groups of six patients each. With free hirudin in 20- to 10,000-fold molar excess of thrombin and peak activated partial thromboplastin times of 2.3 to 3.0 times baseline, mean plasma thrombin-hirudin complex increased from 794 +/- 85 pg/mL (mean +/- SEM) 15 minutes after the start of the infusion to 1617 +/- 151 pg/mL at 6 hours of infusion to 2667 +/- 654 pg/mL at 24 hours. During the 24-hour observation period, plasma concentration of fragment 1.2 (the peptide released during conversion of prothrombin to thrombin) never fell below baseline but rather increased transiently during the hirudin infusion. Plasma concentrations of thrombin-antithrombin III complex (in ng/mL) decreased from 4.34 +/- 0.40 at baseline to 1.64 +/- 0.13 at 6 hours (P < .001) and gradually increased after stopping the infusion to 5.7 +/- 0.87 at 24 hours (nonsignificant compared with baseline). CONCLUSIONS: Measurement of thrombin-hirudin complex may be used as a marker of thrombin generation in humans. Persistent accumulation of thrombin-hirudin complex and generation of fragment 1.2 during and after completion of potent anticoagulation with hirudin suggest thrombin generation is not blocked by high-affinity thrombin inhibition. The persistent formation of thrombin during declining plasma levels of hirudin may contribute to the pathogenesis of rethrombosis early after antithrombin therapy or during inadequate anticoagulation.


Subject(s)
Hirudins/pharmacology , Thrombin/antagonists & inhibitors , Thrombin/biosynthesis , Aged , Antithrombin III/analysis , Enzyme-Linked Immunosorbent Assay , Hirudins/metabolism , Humans , Male , Middle Aged , Osmolar Concentration , Peptide Fragments/analysis , Peptide Hydrolases/analysis , Prothrombin/analysis , Thrombin/metabolism
18.
Tex Heart Inst J ; 21(1): 98-103, 1994.
Article in English | MEDLINE | ID: mdl-8180517

ABSTRACT

Gene transfer by virus- and liposome-mediated vectors has potential for treating genetic diseases, cancer, and cardiovascular diseases. In this article, we discuss the general principle and techniques for gene transfer and the specific issues facing therapy for vascular diseases. We also propose a strategy for using virus-mediated gene transfer to restore the vasoprotective function of the vascular wall, thereby preventing vascular thrombosis. Experimental data from ongoing work in our laboratories are presented to illustrate the importance of this approach in vascular gene transfer therapy.


Subject(s)
Genetic Therapy , Vascular Diseases/therapy , Gene Transfer Techniques , Genetic Vectors , Humans
19.
Circulation ; 88(5 Pt 1): 2015-22, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8222093

ABSTRACT

BACKGROUND: Because the specific antithrombin hirudin prevents platelet-rich arterial thrombus and accelerates thrombolysis in a variety of animal models, it has promise as antithrombotic therapy. We therefore studied the half-life, effect on anticoagulant parameters, and safety of hirudin in patients with coronary artery disease. METHODS AND RESULTS: Thirty-eight men and 1 woman (age [mean +/- SD], 60.4 +/- 6.9 years) with angiographic coronary disease were allocated in a single-blind ascending dosage study to a 6-hour i.v. infusion of recombinant hirudin (CGP 39,393) or matching placebo. The median terminal half-life for hirudin, measured by ELISA, was 2.7, 2.3, 2.9, 3.1, and 2.0 hours for the 0.02, 0.05, 0.1, 0.2, and 0.3 mg.kg-1 x h-1 groups, respectively. Activated partial thromboplastin times (aPTT) at 3, 4, and 6 hours were averaged into a plateau value. The aPTT plateau-to-baseline ratios were 1.5 +/- 0.1, 2.0 +/- 0.1, 2.3 +/- 0.1, 2.7 +/- 0.1, and 2.9 +/- 0.1, respectively, with hirudin infused at 0.02, 0.05, 0.1, 0.2, and 0.3 mg.kg-1 x h-1. From 62% to 77% of the aPTT plateau value was seen within 30 minutes of starting the infusions and was directly related to dose. The aPTT-to-baseline ratios correlated well with plasma hirudin levels (r = .88), whereas poor correlation and sensitivity were observed between plasma hirudin levels and activated coagulation time (ACT)-to-baseline ratios (r = .44). Plasma levels of hirudin and ACT in seconds correlated overall well (r = .80), but considerable overlap occurred between baseline ACT and ACT at plasma hirudin concentrations < 1000 ng/mL. Prothrombin times were significantly prolonged only at a dosage of > or = 0.05 mg.kg-1 x h-1 and were 11.8 +/- 0.5 (INR = 1.0), 12.3 +/- 0.7 (INR = 1.1), 13.3 +/- 1.2 (INR = 1.4), 14.2 +/- 0.4 (INR = 1.7), and 15.8 +/- 0.9 (INR = 2.3) seconds for each respective hirudin dosage. Thrombin times were beyond range (> 600 seconds) at 6 hours in all except 2 patients who received the lowest dosage. All parameters returned to baseline between 8 and 18 hours after the infusion. Bleeding times were not significantly prolonged. No side effects occurred. No antibodies to hirudin were detected 2 weeks after the infusion. CONCLUSIONS: Recombinant hirudin has a terminal half-life of 2 to 3 hours. The aPTT correlates well with plasma levels of hirudin and allows close titration over a wide range of anticoagulation, while ACT and prothrombin time are relatively insensitive for monitoring hirudin administration. At anticoagulant levels effective in experimental thrombosis, a 6-hour infusion of hirudin is well tolerated and safe in a predominantly male group of patients with stable coronary atherosclerosis.


Subject(s)
Blood Coagulation/drug effects , Coronary Disease/etiology , Hirudin Therapy , Aged , Bleeding Time , Blood Coagulation Tests , Chronic Disease , Coronary Disease/physiopathology , Dose-Response Relationship, Drug , Female , Half-Life , Hirudins/adverse effects , Hirudins/blood , Humans , Male , Middle Aged , Recombinant Proteins
20.
Proc Natl Acad Sci U S A ; 90(5): 1819-23, 1993 Mar 01.
Article in English | MEDLINE | ID: mdl-8446595

ABSTRACT

The amount of thrombin active in vivo in the intravascular space (blood and endothelial surface), both basally and in experimental intravascular coagulation, is measured by way of the accessibility of thrombin to intravascular hirudin. Blood samples from pigs given intravenous 125I-labeled hirudin contain 125I-labeled hirudin-thrombin complex in concentrations indicative of a basal thrombin concentration in vivo of 0.5 nmol/liter. Intravenous infusion of Salmonella endotoxin elicits an increase in the circulating concentration of hirudin-thrombin complex that begins within 15 min and is 20-30 times basal after 4 hr. Induction of mild intravascular coagulation is evidenced by a modest reduction in plasma fibrinogen concentrations. It is concluded that there is a basal pool of hirudin-accessible thrombin in the intravascular space that, were it free in the plasma phase, would be sufficient in principle to sustain intravascular coagulation.


Subject(s)
Blood Coagulation , Hirudins , Thrombin/metabolism , Animals , Calcium/metabolism , Disseminated Intravascular Coagulation/metabolism , Endothelium, Vascular/metabolism , Extracellular Space/metabolism , Hirudins/pharmacokinetics , Metabolic Clearance Rate , Protein Binding , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Species Specificity , Swine , Thrombin/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL