Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 162(8): 1855-70, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21232034

ABSTRACT

BACKGROUND AND PURPOSE: rhuMAb Beta7 is a humanized anti-human ß7 monoclonal antibody currently in phase I in inflammatory bowel disease. rhuMAb Beta7 binds the ß7 subunit of the integrins α4ß7 and αEß7, blocking interaction with their ligands. These integrins play key roles in immune cell homing to and retention in mucosal sites, and are associated with chronic inflammatory diseases of the gastrointestinal tract. The goal of this study was to evaluate the mucosal specificity of rhuMAb Beta7. EXPERIMENTAL APPROACH: We assessed the effect of murine anti-Beta7 on lymphocyte homing in mouse models of autoimmune disease. We also compared the effect of rhuMAb Beta7 on circulating mucosal-homing versus peripheral-homing T cells in naïve non-human primates. KEY RESULTS: In cynomolgus monkeys, occupancy of ß7 integrin receptors by rhuMAb Beta7 correlated with an increase in circulating ß7(+) mucosal-homing lymphocytes, with no apparent effect on levels of circulating ß7(-) peripheral-homing lymphocytes. rhuMAb Beta7 also inhibited lymphocyte homing to the inflamed colons of severe combined immunodeficient mice in CD45RB(high) CD4(+) T-cell transfer models. Consistent with a lack of effect on peripheral homing, in a mouse model of experimental autoimmune encephalomyelitis, anti-ß7 treatment resulted in no amelioration of CNS inflammation. CONCLUSIONS AND IMPLICATIONS: The results presented here suggest that rhuMAb Beta7 selectively blocks lymphocyte homing to the gastrointestinal tract without affecting lymphocyte trafficking to non-mucosal tissues. rhuMAb Beta7 provides a targeted therapeutic approach with the potential for a more attractive benefit:risk ratio than currently available inflammatory bowel disease therapies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Integrin beta Chains/immunology , T-Lymphocytes/metabolism , Animals , Antibodies, Monoclonal, Humanized , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/immunology , Colitis/drug therapy , Colitis/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Inflammation/drug therapy , Inflammation/immunology , Intestinal Mucosa/immunology , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Receptors, Lymphocyte Homing/immunology , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/immunology
2.
Thromb Haemost ; 85(3): 475-81, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11307818

ABSTRACT

The substrate recognition region of tissue factor contains two residues, Lys165 and Lys166, which are important for macromolecular substrate activation by the tissue factor:factor VIIa complex. Replacement of these two residues with alanine in a soluble version of human tissue factor resulted in a mutant, hTFAA, which can bind factor VIIa but forms an enzymatically inactive complex. We found that hTFAA inhibits the activity of guinea pig factor VIIa, allowing us to evaluate hTFAA's effects on thrombosis and hemostasis in a guinea pig model of recurrent arterial thrombosis. In addition to heparin, the effects of hTFAA were compared to active site inhibited factor IXa (F.IXai) and factor Xa (F.Xai). We found that hTFAA, F.IXai and F.Xai were potent antithrombotics and may possess a decreased risk of hemorrhage when compared to unfractionated heparin. When administered at a dose that inhibited thrombosis by about 90%, hTFAA neither affected cuticle bleeding nor the activated partial thromboplastin time, and had only a modest effect on the prothrombin time. At equi-efficacious doses, F.IXai, F.Xai and heparin prolonged bleeding times by 20% (p >0.5), 50% (p <0.05) and 100% (p <0.01), respectively. In summary, our study demonstrates that, unlike heparin, specific inhibitors of factors VIIa, IXa and Xa can produce antithrombotic effects without or with only minimally disturbing normal hemostasis. The results further suggest that factor VIIa and factor IXa are especially promising targets for antithrombotic drug development.


Subject(s)
Thromboplastin/genetics , Thrombosis/prevention & control , Amino Acid Substitution , Animals , Arterial Occlusive Diseases/drug therapy , Arterial Occlusive Diseases/etiology , Arterial Occlusive Diseases/prevention & control , Bleeding Time , Carotid Artery Thrombosis/drug therapy , Carotid Artery Thrombosis/etiology , Carotid Artery Thrombosis/prevention & control , Catalytic Domain , Disease Models, Animal , Factor IXa/antagonists & inhibitors , Factor IXa/pharmacology , Factor VIIa/drug effects , Factor VIIa/metabolism , Factor Xa/pharmacology , Factor Xa Inhibitors , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/pharmacology , Guinea Pigs , Heparin/administration & dosage , Heparin/pharmacology , Humans , Solubility , Thromboplastin/administration & dosage , Thromboplastin/pharmacology , Thrombosis/drug therapy , Thrombosis/etiology
3.
Thromb Haemost ; 82(3): 1188-95, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10494786

ABSTRACT

10C12, a human antibody F(ab')2, which specifically binds to the Gla domain of factor IX, interfered with all known coagulation processes that involve factor IX/IXa. These include the function of the intrinsic Xase complex and the activation of zymogen factor IX by factor XIa and by the tissue factor:factor VIla complex. Furthermore, 10C12 potently inhibited activated partial thromboplastin clotting times (APTT) in plasma of guinea pig and rat, thus enabling in-vivo evaluation. In guinea pigs, a bolus administration of 10C12 (10 microg/kg) prevented cyclic flow variations in damaged carotid arteries without affecting coagulation or bleeding parameters. At a 100-fold higher dose, 10C12 had no effect on normal hemostasis as assessed by the cuticle bleeding time. At this dose, 10C12 was also efficacious in a rat arterial thrombosis model, substantially reducing clot weight and duration of vessel occlusion while prolonging ex-vivo APTT only 1.2-fold. The dose of heparin required to produce comparable antithrombotic effects prolonged the APTT by 12-fold and increased the tail bleeding time (TBT) by 8-fold. In contrast, 10C12 had no effect on TBT. However, rat tails showed a tendency for rebleeding which 10C12 exacerbated. In conclusion, the antithrombotic potency of the 10C12 antibody in two species provides evidence for an important role of F.IX, and its Gla domain in particular, during thrombogenesis under arterial flow conditions. The relative safety at effective doses of this fully human antibody suggests that it may have therapeutic value for treatment of thrombotic disorders.


Subject(s)
1-Carboxyglutamic Acid/immunology , Factor IX/immunology , Factor IX/metabolism , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/pharmacology , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fab Fragments/pharmacology , Animals , Binding Sites , Blood Coagulation/drug effects , Disease Models, Animal , Factor IX/antagonists & inhibitors , Guinea Pigs , Hemostasis/drug effects , Heparin/pharmacology , Humans , Male , Rats , Rats, Sprague-Dawley , Thrombosis/blood , Thrombosis/therapy
4.
Thromb Haemost ; 79(1): 169-76, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9459344

ABSTRACT

Ro 44-3888 is a potent and selective antagonist of GP IIb/IIIa. Following I.V. administration to rhesus monkeys, the (mean +/- SD.) clearance, volume of distribution and terminal half-life of Ro 44-3888 were 4.4 +/- 1.8 ml/min/kg, 0.8 +/- 0.4 l/kg and 2.5 +/- 0.8 h respectively. Oral administration of Ro 48-3657 (1 mg/kg), a doubly protected prodrug form, produced peak concentrations of Ro 44-3888 (152 +/- 51 ng/ml), 4.2 +/- 2.2 h after dosing. Terminal half-life and estimated bioavailability were 5.1 +/- 1.6 h and 33 +/- 6% respectively. No effect on blood pressure, heart rate or platelet counts were seen. Adenosine diphosohate (ADP) induced platelet aggregation (PA) and cutaneous bleeding times (CBT) were determined prior to and after the last of 8 daily oral administrations of Ro 48-3657 (0.25 or 0.5 mg/kg) to eight rhesus monkeys. Peak and trough plasma concentrations were proportional to dose and steady state was achieved after the second administration. Inhibition of PA and prolongation of CBT were concentration dependent. The ex vivo IC50 (82 nM) for ADP-mediated PA correlated with a value (58 nM) determined in vitro. The CBT response curve was displaced to the right of the PA curve. CBT was prolonged to > or = 25 min when levels of Ro 44-3888 exceeded 190 nM and PA was > 90% inhibited. Therefore, in rhesus monkeys, Ro 48-3657 is reproducibly absorbed and converted to its active form, is well tolerated, and has a concentration-dependent effect on PA and CBT. These properties make Ro 48-3657 an attractive candidate for evaluation in patients at high risk for arterial thrombosis.


Subject(s)
Amidines/pharmacology , Piperidines/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Prodrugs/pharmacokinetics , Administration, Oral , Amidines/adverse effects , Amidines/pharmacokinetics , Animals , Drug Administration Schedule , Hematologic Tests , Macaca mulatta , Oximes/pharmacology , Piperidines/adverse effects , Piperidines/pharmacokinetics , Prodrugs/adverse effects , Prodrugs/pharmacology
5.
Blood ; 89(9): 3219-27, 1997 May 01.
Article in English | MEDLINE | ID: mdl-9129026

ABSTRACT

One approach to developing safer and more efficacious agents for the treatment of thrombotic disease involves the design and testing of inhibitors that block specific steps in the coagulation cascade. We describe here the development of a mutant of human tissue factor (TF) as a specific antagonist of the extrinsic pathway of blood coagulation and the testing of this mutant in a rabbit model of arterial thrombosis. Alanine substitutions of Lys residues 165 and 166 in human TF have been shown previously to diminish the cofactor function of TF in support of factor X (FX) activation catalyzed by factor VIIa (FVIIa). The K165A:K166A mutations have been incorporated into soluble TF (sTF; residues 1-219) to generate the molecule "hTFAA." hTFAA binds FVIIa with kinetics and affinity equivalent to wild-type sTF, but the hTFAA x FVIIa complex shows a 34-fold reduction in catalytic efficiency for FX activation relative to the activity measured for sTF x FVIIa. hTFAA inhibits the activation of FX catalyzed by the complex formed between FVIIa and relipidated TF(1-243). hTFAA prolongs prothrombin time (PT) determined with human plasma and relipidated TF(1-243) or membrane bound TF, and has no effect on activated partial thromboplastin time, but is 70-fold less potent as an inhibitor of PT with rabbit plasma. The rabbit homologue of this mutant ("rTFAA") was produced and shown to have greater potency with rabbit plasma. Both hTFAA and rTFAA display an antithrombotic effect in a rabbit model of arterial thrombosis with rTFAA giving full efficacy at a lower dose than hTFAA. Compared to heparin doses of equal antithrombotic potential, hTFAA and rTFAA cause less bleeding as judged by measurements of the cuticle bleeding time. These results indicate that TF x FVIIa is a good target for the development of new anticoagulant drugs for the treatment of thrombotic disease.


Subject(s)
Anticoagulants/pharmacology , Fibrinolytic Agents/pharmacology , Partial Thromboplastin Time , Prothrombin Time , Thromboplastin/pharmacology , Animals , Brain/metabolism , Carotid Arteries/drug effects , Carotid Arteries/pathology , Cell Line , Cloning, Molecular , Coagulants/pharmacology , DNA Primers , Escherichia coli , Factor VIIa/metabolism , Heparin/pharmacology , Humans , Kinetics , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Solubility , Thromboplastin/biosynthesis , Thromboplastin/isolation & purification
6.
J Med Chem ; 39(16): 3139-47, 1996 Aug 02.
Article in English | MEDLINE | ID: mdl-8759635

ABSTRACT

The potent and selective GP IIb-IIIa antagonist lamifiban (1, Ro 44-9883) is currently in clinical development as an injectable antithrombotic agent for treating and preventing acute coronary syndromes. However, for secondary prevention of thrombotic occlusions, orally active inhibitors are needed. By means of a prodrug strategy, the modest oral absorption of 1 in mice was improved by a factor of 9. In addition, these studies demonstrated that an amidoxime group can serve as a prodrug functionality for an amidino group. Application of this principle to the structurally related amidino carboxylate 13 led to the amidoxime ester 18 which was absorbed approximately 20 times better, after oral administration to mice, than 13. Due to the modification of the amidino group as well as of the carboxylate group, 18 completely lost its ability to interact with purified platelet GP IIb-IIIa. After oral administration of 18 to rats, dogs, and rhesus monkeys, the bioavailability of the active derivative 13 was 26 +/- 5, 25 +/- 6, and 33 +/- 6%, respectively, and the elimination half-life was 4.1 +/- 1.7, 11.4 +/- 1.1, and 5.1 +/- 1.4 h, respectively. On the basis of these properties, the orally active 18 (Ro 48-3657), a double prodrug of the potent and selective non-peptide GP IIb-IIIa antagonist 13 (Ro 44-3888), was selected as clinical candidate for evaluation as a prophylactic agent in patients at high risk for arterial thrombosis.


Subject(s)
Amidines/pharmacology , Oximes/pharmacology , Piperidines/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Prodrugs/pharmacology , Acetates/pharmacology , Administration, Oral , Amidines/chemistry , Amidines/pharmacokinetics , Animals , Anticoagulants/administration & dosage , Anticoagulants/chemical synthesis , Anticoagulants/pharmacokinetics , Anticoagulants/pharmacology , Dogs , Macaca mulatta , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Molecular Structure , Oximes/chemistry , Oximes/pharmacokinetics , Piperidines/chemistry , Piperidines/pharmacokinetics , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/pharmacokinetics , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Rats , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
7.
Thromb Haemost ; 75(6): 915-20, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8822586

ABSTRACT

Clinical experience suggests that thrombolytic-induced bleeding is associated with systemic activation of the thrombolytic system. Using fibrin specific variants of tissue-type plasminogen activator (t-PA) and making use of the apparent fibrin specificity of streptokinase (SK) in the rabbit we tested the hypothesis that minimizing systemic plasmin production and fibrinogenolysis will decrease hemorrhages in models of peripheral bleeding and embolic stroke. t-PA consumed 51% of the available fibrinogen; caused cerebral bleeds and increased peripheral bleeding time. Fibrin-specific variants of t-PA depleted less than 20% of the fibrinogen and did not cause peripheral or cerebral bleeding. However, an equipotent dose of SK converted only 12% of the available fibrinogen but increased bleeding time and caused hemorrhagic conversion in 75% of embolic stroke model animals treated. The data suggest that bleeding associated with tissue-type plasminogen activators is linked to systemic plasmin generation and subsequent fibrinogenolysis. This hypothesis does not explain the mechanism(s) of SK-induced bleeding.


Subject(s)
Hemorrhage/blood , Plasminogen/metabolism , Streptokinase/therapeutic use , Tissue Plasminogen Activator/blood , Animals , Bleeding Time , Fibrin/metabolism , Hemorrhage/prevention & control , Rabbits , Streptokinase/metabolism
8.
Thromb Haemost ; 75(5): 820-6, 1996 May.
Article in English | MEDLINE | ID: mdl-8725730

ABSTRACT

Multiple clinical trials have proven that thrombolytic therapy is an effective treatment for acute myocardial infarction. Spontaneous intracranial hemorrhage (ICH) occurs in a small percentage of patients as a result of the treatment. The etiology of the ICH is unknown and there is currently no established experimental model for this side effect. A model of ICH during thrombolytic therapy has been developed using spontaneously hypertensive rats (SHR). The SHR were made susceptible to ICH during thrombolytic therapy by bilateral ligation of the external jugular veins. This procedure produced asymptomatic hemorrhagic lesions in the brains of the animals in the hours preceding the administration of t-PA/heparin. The incidence of ICH following the administration of test substances was assessed by histological examination and by measuring the red blood cell count in a sample of cerebrospinal fluid taken from the atlanto-occipital space. t-PA administration produced a low frequency of ICH in this model. The incidence and severity of ICH were dramatically increased, and significant mortality at 24h was observed, by combining heparin were administered sequentially rather than simultaneously. Furthermore, ICHs were observed whether the t-PA dose was administered over 4 h, 1 h, or as a double bolus 30 min apart. The potentiation of ICH by heparin was dose dependent and proportional to the prolongation of the aPTT. Although the precise mechanism of ICH during thrombolytic therapy is unknown, many similarities exist between the observations made in this model and in the human clinical experience.


Subject(s)
Cerebral Hemorrhage , Disease Models, Animal , Fibrinolytic Agents/administration & dosage , Hypertension/complications , Streptokinase/administration & dosage , Tissue Plasminogen Activator/administration & dosage , Animals , Antithrombin III/administration & dosage , Cerebral Hemorrhage/etiology , Drug Interactions , Heparin/pharmacology , Hirudins/administration & dosage , Humans , Male , Rats , Rats, Inbred SHR
9.
Circulation ; 92(10): 3032-40, 1995 Nov 15.
Article in English | MEDLINE | ID: mdl-7586274

ABSTRACT

BACKGROUND: The thrombolytic properties of a new variant of tissue plasminogen activator (TPA) (T103N, N117Q, KHRR 296-299 AAAA, or TNK-TPA) with longer plasma half-life, greater fibrin specificity, and increased resistance to inhibition by plasminogen activator inhibitor (PAI-1) were investigated in a rabbit thrombosed carotid artery model. METHODS AND RESULTS: After 60 minutes of arterial occlusion, TPA (1.5, 3.0, 6.0, or 9.0 mg/kg as a front-loaded IV infusion for 90 minutes; n = 22) or TNK-TPA (0.38, 0.75, or 1.5 mg/kg as IV bolus; n = 16) was administered. Blood flow through the artery was monitored for an additional 120 minutes. Bleeding was assessed by weighing the amount of blood absorbed in a gauze pad placed in a subcutaneous muscular incision. Recanalization rates and duration of recanalization were dose dependent. The doses that produced > 80% recanalization rates with the longest duration of recanalization were 9.0 mg/kg for TPA and 1.5 mg/kg for TNK-TPA. At these doses, time to reperfusion (mean +/- SEM) was significantly faster (11 +/- 2 versus 23 +/- 7 minutes) and duration of recanalization longer (77 +/- 9 versus 51 +/- 18 minutes) for TNK-TPA compared with TPA (P < .025). Weights of the residual thrombi of the TPA group were greater than those of the TNK-TPA group (P = .004). Concentrations of fibrinogen, plasminogen, and alpha 2-antiplasmin at 120 minutes were significantly higher for TNK-TPA-treated animals compared with TPA-treated animals (P < .001). ANOVA of the blood loss data determined that there were significant differences between thrombolytic agents but not between doses. After correction for saline controls, total blood loss for pooled doses of TPA and TNK-TPA was 82 +/- 6 mg and 40 +/- 4 mg, respectively (P < .01). CONCLUSIONS: From these data, we conclude that TNK-TPA, given as a bolus, produces faster and more complete recanalization of occluded arteries in a rabbit experimental model compared with TPA, without increasing systemic plasmin generation or peripheral bleeding. In addition, we observed that TNK-TPA, unlike TPA, did not potentiate collagen-induced aggregation of platelets obtained from human plasma. This lack of effect on platelet aggregation by TNK-TPA potentially could be associated with a decreased risk of reocclusion after successful thrombolysis.


Subject(s)
Carotid Artery Thrombosis/drug therapy , Fibrinolytic Agents/therapeutic use , Tissue Plasminogen Activator/therapeutic use , Animals , Fibrinolytic Agents/adverse effects , Fibrinolytic Agents/chemistry , Hemorrhage/chemically induced , Humans , Kringles , Male , Molecular Sequence Data , Platelet Aggregation/drug effects , Rabbits , Tissue Plasminogen Activator/adverse effects , Tissue Plasminogen Activator/chemistry
10.
Stroke ; 25(10): 2072-8; discussion 2078-9, 1994 Oct.
Article in English | MEDLINE | ID: mdl-8091454

ABSTRACT

BACKGROUND AND PURPOSE: We compared the activity of a new long-half-life, fibrin-specific tissue-type plasminogen activator (TPA) variant with that of wild-type TPA in rabbit models of embolic stroke and peripheral bleeding. METHODS: In the embolic stroke model. TPA-induced clot lysis is followed by continuous monitoring of a radiolabeled clot lodged in the middle cerebral artery. Twenty-four hours after embolization and treatment with either thrombolytic agent or excipient, the brains are removed, fixed, and evaluated for cerebral hemorrhage. In a parallel template bleeding time experiment, the effects of equipotent doses of the two TPA molecules were measured. RESULTS: Infusion of wild-type TPA or bolus administration of the TPA variant resulted in dose-dependent clot lysis. The TPA variant was found to be an order of magnitude more potent than wild-type TPA on a milligram-per-kilogram basis. Unlike wild-type TPA, the variant caused less systemic activation of plasminogen (P < .05) and fewer hemorrhagic transformations in this model (P < .05). The TPA variant did not extend template bleeding times. CONCLUSIONS: These findings show that by combining increased fibrin specificity with decreased plasma clearance, it is possible to produce a thrombolytic agent that is more convenient and more potent than wild-tpe TPA. At the same time the significant reduction in hemorrhagic conversions may be attributable to the conservation of systemic plasminogen seen with this molecule.


Subject(s)
Cerebrovascular Disorders/drug therapy , Fibrin/metabolism , Hemorrhage/chemically induced , Intracranial Embolism and Thrombosis/drug therapy , Tissue Plasminogen Activator/therapeutic use , Animals , Blood Coagulation/drug effects , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/pathology , Dose-Response Relationship, Drug , Fibrinogen/analysis , Fibrinolysis/drug effects , Half-Life , Intracranial Embolism and Thrombosis/pathology , Male , Plasminogen/analysis , Rabbits , Thrombolytic Therapy , Tissue Plasminogen Activator/administration & dosage , Tissue Plasminogen Activator/adverse effects , Tissue Plasminogen Activator/blood , alpha-2-Antiplasmin/analysis
11.
Thromb Haemost ; 72(1): 98-104, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7974384

ABSTRACT

rt-PA-K, a variant of recombinant tissue-type plasminogen activator (rt-PA) with substitution of amino acids 296 to 299 with alanine (KHRR296-299AAAA) has increased fibrin-specificity and reduced sensitivity to plasminogen activator inhibitor-1; rt-PA-T, with threonine 103 replaced by asparagine has an additional glycosylation site and a reduced clearance; and rt-PA-N, with asparagine 117 mutagenized to glutamine lacks the high mannose carbohydrate side chain. We have investigated whether combination of these properties in a single molecule might yield an improved thrombolytic agent. The thrombolytic potency and fibrin-specificity of the combination mutant rt-PA-TNK was compared with that of rt-PA in a combined venous whole blood clot model and platelet-rich arterial eversion graft thrombosis model in dogs given intravenous heparin and aspirin. Infusion of 0.125 to 1.0 mg/kg over 60 min in groups of 4 to 5 dogs produced dose-dependent fibrin-specific venous clot lysis. The thrombolytic potency (percent lysis per mg compound administered per kg body weight) of rt-PA-TNK was significantly higher than that of rt-PA as evidenced by a higher maximal rate of lysis of 480 +/- 100% versus 140 +/- 40% within the 2 h observation period per mg of compound administered per kg body weight (mean +/- SEM, p = 0.004) and a significantly lower dose of 0.08 +/- 0.01 versus 0.21 +/- 0.04 mg/kg body weight at which the maximal rate of lysis was obtained (p = 0.004).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Femoral Artery , Femoral Vein , Recombinant Proteins/therapeutic use , Thrombolytic Therapy , Thrombosis/drug therapy , Tissue Plasminogen Activator/therapeutic use , Amino Acid Sequence , Animals , Disease Models, Animal , Dogs , Drug Resistance , Glycosylation , Graft Occlusion, Vascular/drug therapy , Humans , Molecular Sequence Data , Plasminogen Activator Inhibitor 1/pharmacology , Platelet Count
12.
Proc Natl Acad Sci U S A ; 91(9): 3670-4, 1994 Apr 26.
Article in English | MEDLINE | ID: mdl-8170967

ABSTRACT

Current treatment with tissue plasminogen activator (tPA) requires an intravenous infusion (1.5-3 h) because the clearance of tPA from the circulation is rapid (t 1/2 approximately 6 min). We have developed a tPA variant, T103N,N117Q, KHRR(296-299)AAAA (TNK-tPA) that has substantially slower in vivo clearance (1.9 vs. 16.1 ml per min per kg for tPA in rabbits) and near-normal fibrin binding and plasma clot lysis activity (87% and 82% compared with wild-type tPA). TNK-tPA exhibits 80-fold higher resistance to plasminogen activator inhibitor 1 than tPA and 14-fold enhanced relative fibrin specificity. In vitro, TNK-tPA is 10-fold more effective at conserving fibrinogen in plasma compared to tPA. Arterial venous shunt models of fibrinolysis in rabbits indicate that TNK-tPA (by bolus) induces 50% lysis in one-third the time required by tPA (by infusion). TNK-tPA is 8- and 13-fold more potent in rabbits than tPA toward whole blood clots and platelet-enriched clots, respectively. TNK-tPA conserves fibrinogen and, because of its slower clearance and normal clot lysis activity, is effective as a thrombolytic agent when given as a bolus at a relatively low dose.


Subject(s)
Fibrinolytic Agents/chemistry , Tissue Plasminogen Activator/chemistry , Animals , Fibrin/metabolism , Fibrinogen/metabolism , Fibrinolysis , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/pharmacokinetics , Humans , Kinetics , Mutagenesis, Site-Directed , Rabbits , Structure-Activity Relationship , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/pharmacokinetics
13.
Thromb Haemost ; 70(2): 307-12, 1993 Aug 02.
Article in English | MEDLINE | ID: mdl-8236140

ABSTRACT

Site directed mutagenesis was used to construct a t-PA variant that contains an additional glycosylation site in the first kringle domain (T103N) combined with a tetra-alanine substitution in the protease domain (KHRR 296-299 AAAA). This combination variant has a plasma clearance rate that is 4.5-fold slower in rats and 5.4-fold slower in rabbits than t-PA. It is also less than one tenth as active as t-PA towards plasminogen in the presence of fibrinogen, and has approximately twice the normal activity in the presence of fibrin. It shows substantial resistance to the fast acting inhibitor, plasminogen activator inhibitor-1 (PAI-1), requiring a 10-fold greater molar excess of PAI-1 to reduce its activity by 50%, compared to t-PA. This is the result of a reduction of nearly 100-fold in the second order rate constant for PAI-1 inactivation. These results show that it is possible to combine mutations in different domains of t-PA to construct a variant which is simultaneously slower clearing, less reactive towards plasminogen in the absence of a fibrin clot, and resistant to inactivation by PAI-1.


Subject(s)
Plasminogen Activator Inhibitor 1/pharmacology , Tissue Plasminogen Activator/pharmacokinetics , Animals , CHO Cells , Cricetinae , Fibrin/metabolism , Metabolic Clearance Rate , Mutagenesis, Site-Directed , Plasminogen Activator Inhibitor 1/metabolism , Protein Structure, Tertiary , Rabbits , Rats , Substrate Specificity , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/metabolism
14.
Thromb Haemost ; 70(2): 313-9, 1993 Aug 02.
Article in English | MEDLINE | ID: mdl-8236141

ABSTRACT

In the accompanying paper, we reported that the properties of decreased plasma clearance rate, increased fibrin specificity, and resistance to inactivation by PAI-1 could be effectively combined in the t-PA variant T103N, KHRR 296-299 AAAA. In the current study we evaluated the in vivo efficacy of this variant as well as variants containing the individual mutations T103N and KHRR 296-299 AAAA. Plasma clearance and in vivo lysis of whole blood and platelet-rich clots were determined in a rabbit arterio-venous shunt model. The T103N containing variants were administered as an intravenous (i.v.) bolus. KHRR 296-299 AAAA and t-PA were infused i.v. over 90 min. The clearance rate of the KHRR 296-299 AAAA variant was similar to t-PA. However, the clearance of the T103N and T103N, KHRR 296-299 AAAA variants were 8 and 6-fold reduced, respectively. Potency of the variants relative to t-PA on whole blood clots ranged from 0.9 (T103N, KHRR 296-299 AAAA) to 1.7 (T103N). Relative potency on platelet-rich clots ranged from 2.4 (T103N) to 4.2 (T103N, KHRR 296-299 AAAA). Fibrinogen concentrations in rabbits 120 min after dosing with a 2.5 mg/kg bolus were: 24, 16, 82, and 77% of initial for t-PA; T103N; KHRR 296-299 AAAA; and T103N, KHRR 296-299 AAAA treatment groups, respectively. These results suggest that the T103N, KHRR 296-299 AAAA variant of t-PA, given as a bolus, could result in greater efficacy, particularly on refractory platelet-rich clots, without inducing the severe systemic lytic state produced by a bolus of a less fibrin specific variant.


Subject(s)
Fibrinolytic Agents/therapeutic use , Thrombolytic Therapy , Tissue Plasminogen Activator/pharmacology , Animals , Arteriovenous Shunt, Surgical , Disease Models, Animal , Dose-Response Relationship, Drug , Injections, Intravenous , Metabolic Clearance Rate , Platelet Aggregation , Rabbits , Recombinant Proteins/pharmacology , Tissue Plasminogen Activator/administration & dosage , Tissue Plasminogen Activator/pharmacokinetics
15.
J Pharmacol Exp Ther ; 264(1): 67-73, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8423552

ABSTRACT

The dose-dependent effects of tissue-type plasminogen activator (t-PA) on the kinetics of cerebral clot lysis in a rabbit model of middle cerebral artery embolic stroke were investigated. The clots were formed in vitro and tagged with 99Tc for gamma-scintigraphic imaging. After embolization, groups of animals were treated with t-PA. Dose-response curves for the t-PA were generated, and in addition, long and short dosing schedules were assessed. The optimal doses for frequency and rate of cerebral clot lysis in this model are approximately 6.3 mg/kg given over 2 hr or 3.3 mg/kg given over 30 min. These dosing regimens for t-PA were accompanied by approximately 50% consumption of plasma plasminogen, fibrinogen and alpha 2-antiplasmin. Doses of t-PA on either side of this optimum caused attenuation in both the frequency and rate of cerebral clot lysis. Treatment with t-PA under either dosing regimen did not augment the frequency of hemorrhagic transformation, but the size of the resultant hemorrhage in those animals where intracranial bleeding occurred was reduced by 3.3 mg/kg t-PA given over 30 min.


Subject(s)
Intracranial Embolism and Thrombosis/drug therapy , Thrombolytic Therapy , Tissue Plasminogen Activator/therapeutic use , Animals , Cerebral Arteries , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Intracranial Embolism and Thrombosis/complications , Male , Rabbits , Technetium Tc 99m Sulfur Colloid
16.
Protein Eng ; 5(3): 259-66, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1409547

ABSTRACT

The tetra-alanine substitution variant KHRR 296-299 AAAA of tissue-type plasminogen activator (t-PA) was previously shown to have enhanced fibrin specificity and enhanced activity in the presence of fibrin compared with the wild-type form of the molecule. The structural requirements for these alterations in enzymatic activity were investigated by constructing several amino acid substitution variants at each of the positions from 296 to 299 and evaluating their activities under a variety of conditions. Effects on plasminogen activator activity were common among the point mutants at positions 296-299; nearly all had a phenotype similar to the KHRR 296-299 AAAA variant. The greatest effects on enzymatic function were found with multiple substitution variants, but some single charge reversals and proline substitutions had substantial effects. The enhanced fibrin specificity of KHRR 296-299 AAAA t-PA results in less fibrinogenolysis than seen with wild-type t-PA. Approximately four times greater concentration of KHRR 296-299 AAAA compared with wild-type t-PA was required to consume 50% of the fibrinogen in human plasma.


Subject(s)
Alanine/chemistry , Tissue Plasminogen Activator/metabolism , Amino Acid Sequence , Fibrin/metabolism , Fibrinogen/metabolism , Fibrinolysis , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligopeptides/metabolism , Plasma , Plasminogen/metabolism , Protein Engineering , Tissue Plasminogen Activator/chemistry
17.
Thromb Haemost ; 60(2): 255-61, 1988 Oct 31.
Article in English | MEDLINE | ID: mdl-2851193

ABSTRACT

Modification of the carbohydrate structures of recombinant tissue-type plasminogen activator (rt-PA) can increase or decrease its rate of clearance in rabbits. When rt-PA was treated with sodium periodate to oxidize carbohydrate residues, the rate of clearance was decreased from 9.6 +/- 1.9 ml min-1 kg-1 to 3.5 +/- 0.6 ml min-1 kg-1 (mean +/- SD, n = 5). A similar change in the clearance of rt-PA was introduced by the use of endo-beta-N-acetyl-glucosaminidase H (Endo-H), which selectively removes high mannose asparagine-linked oligosaccharides; the clearance of Endo-H-treated rt-PA was 5.0 +/- 0.5 ml min-1 kg-1. A mutant of rt-PA was produced with an amino acid substitution at position 117 (Asn replaced with Gln) to remove a potential glycosylation site that normally contains a high mannose structure. The clearance of this material was also decreased, similar to the periodate and Endo-H-treated rt-PA. Conversely, when rt-PA was produced in the CHO 15B cell line, which can produce only high mannose oligosaccharide structures on glycoproteins, the clearance was increased by a factor of 1.8. These results demonstrate that the removal of rt-PA from the blood depends significantly upon the nature of its oligosaccharide structures.


Subject(s)
Carbohydrates , Tissue Plasminogen Activator/pharmacokinetics , Animals , Cells, Cultured , Cricetinae , Glutamine/analysis , Mannose/analysis , Mutation , Oligosaccharides/analysis , Periodic Acid/pharmacology , Rabbits , Recombinant Proteins/pharmacokinetics
18.
Thromb Haemost ; 56(2): 160-4, 1986 Oct 21.
Article in English | MEDLINE | ID: mdl-2433785

ABSTRACT

In vitro artifacts due to proteolysis may occur in blood samples containing recombinant tissue-type plasminogen activator (rt-PA) due to continued activation of plasminogen to plasmin by rt-PA. The aim of this study was to identify a rapid inhibitor of rt-PA that would not interfere in assays designed to monitor thrombolytic events. When rt-PA was added at 5 micrograms/ml to whole blood and incubated at 25 degrees C, fibrinogen decreased 50 percent, plasminogen levels decreased 90 percent and alpha 2-antiplasmin decreased below detectable levels. If D-Phe-Pro-Arg-chloromethylketone (PPACK) or aprotinin were added before the addition of rt-PA there was no significant loss of fibrinogen. Only PPACK completely inhibited changes in fibrin degradation products, plasminogen and alpha 2-antiplasmin. PPACK was also found to inhibit the binding of rt-PA to plasma protease inhibitors in vitro. Rhesus monkeys were infused with rt-PA and blood samples were taken with either PPACK or aprotinin in the collection syringe. There was a significant increase in the recovery of immunoreactive rt-PA and consistent measures of fibrinogen, FDPs, plasminogen, and alpha 2-antiplasmin in the PPACK group as compared to the aprotinin group which indicates that PPACK will prevent the in vitro formation of artifacts due to the presence of active rt-PA.


Subject(s)
Amino Acid Chloromethyl Ketones/pharmacology , Blood Specimen Collection , Fibrinolytic Agents , Recombinant Proteins/therapeutic use , Thrombin/antagonists & inhibitors , Tissue Plasminogen Activator/therapeutic use , Amino Acid Chloromethyl Ketones/therapeutic use , Aprotinin/pharmacology , Enzyme Activation , Fibrinogen/metabolism , Fibrinolysin/metabolism , Humans , Kinetics , Plasminogen/metabolism
19.
Am J Physiol ; 249(3 Pt 1): E306-11, 1985 Sep.
Article in English | MEDLINE | ID: mdl-2994490

ABSTRACT

The effect of an endurance training regimen on muscle oxidative enzymes and work performance was studied in iron-deficient and -sufficient rats. Three-week-old male Sprague-Dawley rats (n = 40) were randomly assigned to diets containing either 6 mg iron/kg (iron deficient) or 50 mg iron/kg (iron sufficient). After 2 wk, each group of rats was further divided into untrained or endurance-trained subgroups. Training consisted of daily treadmill running of gradually increasing duration for a 1-mo period. After the training period, sedentary and endurance-trained iron-deficient rats were anemic (Hgb approximately 8 g/dl compared with 16 g/dl in the 2 control groups) and had significantly lower skeletal muscle cytochrome c concentration, cytochrome oxidase activity, and succinic oxidase activity compared with the iron-sufficient groups. In response to training iron-deficient rats also generally had a substantial increase in skeletal muscle oxidative enzymes (P less than 0.05), in contrast to iron-sufficient animals, in which there was little or no training effect. Work performance in response to training in the iron-deficient rats improved more than sixfold in an endurance type of exercise (P less than 0.05), but maximal oxygen consumption during a brief, intense type of exercise was not significantly affected. The results suggest that endurance training of iron-deficient rats results in a milder anemia and less drastic reduction of skeletal muscle oxidative enzymes which in turn allows better performance in an endurance type of exercise.


Subject(s)
Iron Deficiencies , Physical Exertion , Animals , Body Weight , Eating , Electron Transport Complex IV/metabolism , Hemoglobins/analysis , Iron/pharmacology , Male , Muscles/anatomy & histology , Muscles/drug effects , Muscles/metabolism , Organ Size/drug effects , Oxygen Consumption/drug effects , Physical Education and Training , Rats , Rats, Inbred Strains , Succinate Dehydrogenase/metabolism , Time Factors
20.
J Appl Physiol (1985) ; 58(5): 1477-80, 1985 May.
Article in English | MEDLINE | ID: mdl-2987175

ABSTRACT

The effect of iron deficiency on work capacity was studied in groups of rats that had received diets with iron contents ranging between 9 and 50 mg/kg diet from 3 to 6 wk of age. Maximal O2 consumption (VO2max) declined only 16% with a decrease in hemoglobin (Hb) from 14 to 8 g/dl and fell sharply only below a Hb of 7 g/dl. Duration until exhaustion in a treadmill exercise of submaximal intensity (endurance) showed no significant depression between a Hb of 14 and 10 g/dl. However, endurance declined abruptly by 73% between a Hb of 10 and 8 g/dl. The VO2max results are in accord with known compensatory mechanisms that help to maintain delivery of O2 to tissues until anemia becomes severe. The sharp fall in endurance with relatively mild iron deficiency suggests a lack of similarly effective compensations for decreased oxidative capacity of muscle.


Subject(s)
Anemia, Hypochromic/physiopathology , Physical Exertion , Anemia, Hypochromic/metabolism , Animals , Cytochrome c Group/metabolism , Diet , Hemoglobins/analysis , Iron/administration & dosage , Lactates/blood , Lactic Acid , Male , Muscle Contraction , Muscles/metabolism , Oxygen Consumption , Physical Endurance , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...