Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 259
Filter
Add more filters

Publication year range
1.
Oncology ; 96(4): 217-222, 2019.
Article in English | MEDLINE | ID: mdl-30844808

ABSTRACT

OBJECTIVES: Tissue factor overexpression is associated with tumor progression, venous thromboembolism, and worsened survival in patients with cancer. Tissue factor and activated factor VII (FVIIa) complex may contribute to tumor invasiveness by promoting cell migration and angiogenesis. The study objective was to evaluate safety, pharmacokinetics, and efficacy of PCI-27483, a selective FVIIa inhibitor. METHODS: This was an open-label, multicenter phase 2 trial of patients with advanced pancreatic cancer. Part A of the study was an intrapatient dose escalation lead-in portion in patients concurrently receiving gemcitabine, and in part B, patients were randomized 1: 1 to the recommended phase 2 dose combination PCI-27483-gemcitabine versus gemcitabine alone. RESULTS: Target international normalized ratio (between 2.0-3.0) was achieved following PCI-27483 treatment. Overall safety of PCI-27483-gemcitabine (n = 26) was similar to gemcitabine alone (n = 16), with a higher incidence of mostly low-grade bleeding events (65% vs. 19%). Progression-free survival (PFS) and overall survival (OS) were not significantly different between patients treated with PCI-27483-gemcitabine (PFS: 3.7 months, OS: 5.7 months) and those treated with gemcitabine alone (PFS: 1.9 months, OS: 5.6 months). CONCLUSIONS: Targeted inhibition of the coagulation cascade was achieved by administering PCI-27483. PCI-27483-gemcitabine was well tolerated, but superiority to single agent gemcitabine was not demonstrated.


Subject(s)
Anticoagulants/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aspartic Acid/analogs & derivatives , Benzimidazoles/administration & dosage , Blood Coagulation/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Factor VIIa/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Aged , Aged, 80 and over , Anticoagulants/adverse effects , Anticoagulants/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Aspartic Acid/administration & dosage , Aspartic Acid/adverse effects , Aspartic Acid/pharmacokinetics , Benzimidazoles/adverse effects , Benzimidazoles/pharmacokinetics , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/secondary , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Drug Monitoring/methods , Factor VIIa/metabolism , Female , Hemorrhage/chemically induced , Humans , International Normalized Ratio , Male , Middle Aged , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Progression-Free Survival , Time Factors , Gemcitabine
2.
Br J Haematol ; 181(4): 528-536, 2018 05.
Article in English | MEDLINE | ID: mdl-29611182

ABSTRACT

Bypassing therapy is essential for the haemostatic management of patients with haemophilia A with inhibitor (PWHA-inh), but the therapeutic effects are inconsistent. We previously reported that activated prothrombin complex concentrates (aPCC) activated factor (F)VIIIin vitro, and was mediated mainly by the activated FVII (FVIIa) contained in aPCC. We have extended those studies to assess global coagulation in whole blood from 18 PWHA-inh in the co-presence of aPCC and FVIII using Ca2+ -triggered rotational thromboelastometry. The clot times (CTs) in the presence of both aPCC (0·05 iu/ml) and recombinant (r)FVIII (1 iu/ml) ex vivo were shortened compared to the aPCC alone (P < 0·01). These enhancing effects of rFVIII were observed, irrespective of recognizing inhibitor epitopes; however, the clot formation time and 'α'-angle were not significantly different. In samples from 7 PWHA-inh post-infusion of aPCC (70-80 iu/kg), only the CTs were shortened in the presence of rFVIIIex vivo compared to its absence (P < 0·05), indicating that the enhanced activity centred on the initiation phase of coagulation. Furthermore, experiments in the co-presence of rFVIIa and rFVIII demonstrated that FVIII accelerated only the CTs. We concluded that FVIII/FVIIa-related coagulation mechanism enhanced global haemostatic function by the co-presence of bypassing agents and FVIII in PWHA-inh.


Subject(s)
Blood Coagulation Factor Inhibitors/blood , Blood Coagulation Factors/administration & dosage , Blood Coagulation/drug effects , Factor VIII , Factor VIIa , Hemophilia A , Blood Coagulation Factors/pharmacokinetics , Factor VIII/administration & dosage , Factor VIII/antagonists & inhibitors , Factor VIII/metabolism , Factor VIIa/antagonists & inhibitors , Factor VIIa/metabolism , Female , Hemophilia A/blood , Hemophilia A/drug therapy , Humans , Male
3.
Bioorg Med Chem Lett ; 27(12): 2650-2654, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28460818

ABSTRACT

Factor VIIa (FVIIa) inhibitors have shown strong antithrombotic efficacy in preclinical thrombosis models with limited bleeding liabilities. Discovery of potent, orally active FVIIa inhibitors has been largely unsuccessful due to the requirement of a basic P1 group to interact with Asp189 in the S1 binding pocket, limiting their membrane permeability. We have combined recently reported neutral P1 binding substituents with a highly optimized macrocyclic chemotype to produce FVIIa inhibitors with low nanomolar potency and enhanced permeability.


Subject(s)
Factor VIIa/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Serine Proteinase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Humans , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/chemistry , Molecular Structure , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship
4.
Biochem Biophys Res Commun ; 470(3): 710-713, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26797521

ABSTRACT

The naturally occurring M358R mutation of the plasma serpin α1-proteinase inhibitor (API) changes both its cleavable reactive centre bond to Arg-Ser and the efficacy with which it inhibits different proteases, reducing the rate of inhibition of neutrophil elastase, and enhancing that of thrombin, factor XIa, and kallikrein, by several orders of magnitude. Although another plasma serpin with an Arg-Ser reactive centre, antithrombin (AT), has been shown to inhibit factor VIIa (FVIIa), no published data are available with respect to FVIIa inhibition by API M358R. Recombinant bacterially-expressed API M358R and plasma-derived AT were therefore compared using gel-based and kinetic assays of FVIIa integrity and activity. Under pseudo-first order conditions of excess serpin over protease, both AT and API M358R formed denaturation-resistant inhibitory complexes with FVIIa in reactions accelerated by TF; AT, but not API M358R, also required heparin for maximal activity. The second order rate constant for heparin-independent API M358R-mediated FVIIa inhibition was determined to be 7.8 ± 0.8 × 10(2) M(-1)sec(-1). We conclude that API M358R inhibits FVIIa by forming inhibitory complexes of the serpin type more rapidly than AT in the absence of heparin. The likely 20-fold excess of API M358R over AT in patient plasma during inflammation raises the possibility that it could contribute to the hemorrhagic tendencies manifested by rare individuals expressing this mutant serpin.


Subject(s)
Blood Coagulation/physiology , Factor VIIa/antagonists & inhibitors , Factor VIIa/metabolism , Thromboplastin/metabolism , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism , Genetic Variation/genetics , Humans , Kinetics , Structure-Activity Relationship
5.
Blood ; 124(23): 3365-72, 2014 Nov 27.
Article in English | MEDLINE | ID: mdl-25428222

ABSTRACT

Treatment of patients with hemophilia A and B has undergone significant advances during the past 2 decades. However, despite these advances, the development of antibodies that inhibit the function of infused clotting factor remains a major challenge and is considered the most significant complication of hemophilia treatment. This chapter reviews current tools available for the care of patients with inhibitors and highlights areas where progress is imminent or strongly needed. For management of bleeding, bypassing agents remain the mainstay of therapy. Recombinant factor VIIa and activated prothrombin complex concentrates are similarly effective in populations of patients with hemophilia and inhibitors; however, individuals may show a better response to one agent over another. Recent studies have shown that prophylaxis with bypassing agents can reduce bleeding episodes by ∼50%-80%. The prophylactic use of bypassing agents is an important tool to reduce morbidity in patients before they undergo immune tolerance induction (ITI) and in those with persistent high titer inhibitors, but cost and lack of convenience remain barriers. Because of the significant burden that inhibitors add to the individual patient and the health care system, inhibitor eradication should be pursued in as many patients as possible. ITI is an effective tool, particularly in patients with severe hemophilia A and good risk profiles, and leads to a return to a normal factor VIII response in ∼60% of patients. However, for the group of patients who fail to respond to ITI or have hemophilia B, new and improved tools are needed.


Subject(s)
Factor VIIa/antagonists & inhibitors , Factor VIIa/therapeutic use , Hemophilia A/therapy , Hemophilia B/therapy , Immune Tolerance , Isoantibodies/immunology , Chemoprevention/methods , Factor VIII/antagonists & inhibitors , Factor VIII/immunology , Factor VIII/therapeutic use , Factor VIIa/immunology , Genetic Therapy/methods , Genetic Therapy/trends , Hemophilia A/genetics , Hemophilia A/immunology , Hemophilia B/genetics , Hemophilia B/immunology , Hemorrhage/prevention & control , Humans , Immune Tolerance/drug effects , Immune Tolerance/genetics , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use
6.
Blood ; 123(17): 2605-13, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24632711

ABSTRACT

The coagulation cascade is activated during viral infections. This response may be part of the host defense system to limit spread of the pathogen. However, excessive activation of the coagulation cascade can be deleterious. In fact, inhibition of the tissue factor/factor VIIa complex reduced mortality in a monkey model of Ebola hemorrhagic fever. Other studies showed that incorporation of tissue factor into the envelope of herpes simplex virus increases infection of endothelial cells and mice. Furthermore, binding of factor X to adenovirus serotype 5 enhances infection of hepatocytes but also increases the activation of the innate immune response to the virus. Coagulation proteases activate protease-activated receptors (PARs). Interestingly, we and others found that PAR1 and PAR2 modulate the immune response to viral infection. For instance, PAR1 positively regulates TLR3-dependent expression of the antiviral protein interferon ß, whereas PAR2 negatively regulates expression during coxsackievirus group B infection. These studies indicate that the coagulation cascade plays multiple roles during viral infections.


Subject(s)
Blood Coagulation , Peptide Hydrolases/metabolism , Virus Diseases/blood , Animals , Dengue Virus/metabolism , Factor VIIa/antagonists & inhibitors , Hepatocytes/metabolism , Humans , Immune System , Immunity, Innate , Macrophages/metabolism , Macrophages/virology , Mice , Models, Biological , Myocarditis/metabolism , Myocarditis/virology , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Receptors, Proteinase-Activated/metabolism , Signal Transduction , Thromboplastin/antagonists & inhibitors , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism
7.
Bioorg Med Chem Lett ; 26(20): 5051-5057, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27612545

ABSTRACT

Selective tissue factor-factor VIIa complex (TF-FVIIa) inhibitors are viewed as promising compounds for treating thrombotic disease. In this contribution, we describe multifaceted exploratory SAR studies of S1'-binding moieties within a macrocyclic chemotype aimed at replacing cyclopropyl sulfone P1' group. Over the course of the optimization efforts, the 1-(1H-tetrazol-5-yl)cyclopropane P1' substituent emerged as an improved alternative, offering increased metabolic stability and lower clearance, while maintaining excellent potency and selectivity.


Subject(s)
Factor VIIa/antagonists & inhibitors , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/pharmacology , Thromboplastin/antagonists & inhibitors , Animals , Dogs , Drug Design , Humans , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacokinetics , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 25(10): 2169-73, 2015.
Article in English | MEDLINE | ID: mdl-25881820

ABSTRACT

Heterocyclic amide isosteres were incorporated into a phenylglycine-based tissue factor/factor VIIa (TF-FVIIa) inhibitor chemotype, providing potent inhibitors. An X-ray co-crystal structure of phenylimidazole 19 suggested that an imidazole nitrogen atom effectively mimics an amide carbonyl, while the phenyl ring forms key hydrophobic interactions with the S1' pocket. Exploration of phenylimidazole substitution led to the discovery of potent, selective and efficacious inhibitors of TF-FVIIa.


Subject(s)
Drug Design , Factor VIIa/antagonists & inhibitors , Imidazoles/chemistry , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/pharmacology , Crystallography, X-Ray , Molecular Structure , Serine Proteinase Inhibitors/chemistry
9.
Mar Drugs ; 14(1): 7, 2015 Dec 30.
Article in English | MEDLINE | ID: mdl-26729138

ABSTRACT

The blood coagulation cascade involves the human coagulation factors thrombin and an activated factor VII (fVIIa). Thrombin and fVIIa are vitamin-K-dependent clotting factors associated with bleeding, bleeding complications and disorders. Thrombin and fVIIa cause excessive bleeding when treated with vitamin-K antagonists. In this research, we explored different strains of toxic Microcystis aeruginosa and cyanobacteria blooms for the probable fVIIa-soluble Tissue Factor (fVIIa-sTF) inhibitors. The algal cells were subjected to acidification, and reverse phase (ODS) chromatography-solid phase extraction eluted by water to 100% MeOH with 20%-MeOH increments except for M. aeruginosa NIES-89, from the National Institute for Environmental Studies (NIES), which was eluted with 5%-MeOH increments as an isolation procedure to separate aeruginosins 89A and B from co-eluting microcystins. The 40%-80% MeOH fractions of the cyanobacterial extract are active against fVIIa-sTF. The fVIIa-sTF active fractions from cultured cyanobacteria and cyanobacteria blooms were subjected to liquid chromatography-mass spectrometry (LC-MS). The 60% MeOH fraction of M. aeruginosa K139 exhibited an m/z 603 [M + H]⁺ attributed to aeruginosin K139, and the 40% MeOH fraction of M. aeruginosa NIES-89 displayed ions with m/z 617 [M - SO3 + H]⁺ and m/z [M + H]⁺ 717, which attributed to aeruginosin 89. Aeruginosins 102A/B and 298A/B were also observed from other toxic strains of M. aeruginosa with positive fVIIa-sTF inhibitory activity. The active fractions contained cyanobacterial peptides of the aeruginosin class as fVIIa-sTF inhibitors detected by LC-MS.


Subject(s)
Anticoagulants/pharmacology , Cyanobacteria/chemistry , Factor VIIa/antagonists & inhibitors , Thromboplastin/antagonists & inhibitors , Chromatography, Liquid , Factor VIIa/drug effects , Fresh Water , Humans , Japan , Mass Spectrometry , Structure-Activity Relationship , Thromboplastin/drug effects
10.
Tohoku J Exp Med ; 237(1): 45-50, 2015 09.
Article in English | MEDLINE | ID: mdl-26346967

ABSTRACT

Ebola virus disease (EVD) has been a great concern worldwide because of its high mortality. EVD usually manifests with fever, diarrhea and vomiting, as well as disseminated intravascular coagulation (DIC). To date, there is neither a licensed Ebola vaccine nor a promising therapeutic agent, although clinical trials are ongoing. For replication inside the cell, Ebola virus (EBOV) must undergo the proteolytic processing of its surface glycoprotein in the endosome by proteases including cathepsin B (CatB), followed by the fusion of the viral membrane and host endosome. Thus, the proteases have been considered as potential targets for drugs against EVD. However, no protease inhibitor has been presented as effective clinical drug against it. A synthetic serine protease inhibitor, nafamostat mesilate (NM), reduced the release of CatB from the rat pancreas. Furthermore, it has anticoagulant activities, such as inhibition of the factor VIIa complex, and has been used for treating DIC in Japan. Thus, NM could be considered as a drug candidate for the treatment of DIC induced by EBOV infection, as well as for the possible CatB-related antiviral action. Moreover, the drug has a history of large-scale production and clinical use, and the issues of safety and logistics might have been cleared. We advocate in vitro and in vivo experiments using active EBOV to examine the activities of NM against the infection and the DIC induced by the infection. In addition, we suggest trials for comparison among anti-DIC drugs including the NM in EVD patients, in parallel with the experiments.


Subject(s)
Antiviral Agents/therapeutic use , Ebolavirus/drug effects , Guanidines/therapeutic use , Hemorrhagic Fever, Ebola/drug therapy , Serine Proteinase Inhibitors/therapeutic use , Animals , Benzamidines , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Ebolavirus/enzymology , Factor VIIa/antagonists & inhibitors , Hemorrhagic Fever, Ebola/complications , Humans , Hyperkalemia/etiology , Hyperkalemia/therapy , Hyponatremia/etiology , Hyponatremia/therapy , Rats , Serine Proteases/metabolism , Virus Replication/drug effects
12.
Haemophilia ; 19(4): 571-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23557542

ABSTRACT

The ONE Registry (OR) was an international prospective observational study of on-demand recombinant factor VIIa (rFVIIa) treatment for mild to moderate bleeds in haemophilia A/B patients with inhibitors. To describe real-world use of single and multi dose rFVIIa and to compare outcomes, including effectiveness, safety, quality of life and treatment satisfaction associated with treatment. Baseline data included demographics, treatment, medical and bleed history and patient/caregiver-reported outcomes regarding bleeds. rFVIIa was prescribed according to routine practice; regimens varied and initial dose was categorized as low (LD, ≤ 120 µg kg(-1) ), intermediate (ID, >120 and <250 µg kg(-1) ) or high (HD, ≥ 250 µg kg(-1) ). OR included 102 patients and 85 (83%) reported 494 bleeds overall. Mean age was 23 years (SD 16.4), with 52% ≥ 18 years. Majority of bleeds (n = 350, 71%) involved ≥ 1 joints; 46% involved a target joint. Median initial dose was 90 µg kg(-1) in LD (range 87-120, n = 156), 174 µg kg(-1) in ID, (range 121-249, n = 127) and 270 µg kg(-1) in HD, (range 250-375, n = 211). For spontaneous bleeds, effective haemostasis rate at 9 h was 63% LD, 60% ID and 56% HD. Rates of combined partially effective/effective haemostasis was 85% LD, 96% ID and 86% HD. Median number of doses in HD was one (range 1-7), compared with two in LD (range 1-17) and ID (range 1-23). No thromboembolic events were reported in 1145 doses given. These observational data in real life are consistent with previous studies which have shown similar overall effectiveness of rFVIIa and similar effectiveness and safety across different patterns of standard initial dosing.


Subject(s)
Factor VIIa/antagonists & inhibitors , Factor VIIa/therapeutic use , Hemophilia A/drug therapy , Internationality , Registries , Demography , Dose-Response Relationship, Drug , Factor VIIa/adverse effects , Hemophilia A/blood , Hemophilia A/complications , Hemorrhage/blood , Hemorrhage/complications , Hemorrhage/drug therapy , Hemostasis/drug effects , Humans , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Treatment Outcome
13.
Bioorg Med Chem Lett ; 23(18): 5239-43, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23927973

ABSTRACT

In this Letter, we describe the synthesis of several nonamidine analogs of biaryl acid factor VIIa inhibitor 1 containing weakly basic or nonbasic P1 groups. 2-Aminoisoquinoline was found to be an excellent surrogate for the benzamidine group (compound 2) wherein potent inhibition of factor VIIa is maintained relative to most other related serine proteases. In an unanticipated result, the m-benzamide P1 (compounds 21a and 21b) proved to be a viable benzamidine replacement, albeit with a 20-40 fold loss in potency against factor VIIa.


Subject(s)
Carboxylic Acids/chemistry , Drug Discovery , Factor VIIa/antagonists & inhibitors , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Benzamidines , Crystallography, X-Ray , Dose-Response Relationship, Drug , Factor VIIa/metabolism , Humans , Models, Molecular , Molecular Structure , Serine Proteinase Inhibitors/chemical synthesis , Structure-Activity Relationship
14.
Bioorg Med Chem Lett ; 23(8): 2432-5, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23478148

ABSTRACT

A 6-amidinotetrahydroquinoline screening hit was driven to a structurally novel, potent, and selective FVIIa inhibitor through a combination of library synthesis and rational design. An efficient gram-scale synthesis of the active enantiomer BMS-593214 was developed, which required significant optimization of the key Povarov annulation. Importantly, BMS-593214 showed antithrombotic efficacy in a rabbit arterial thrombosis model. A crystal structure of BMS-593214 bound to FVIIa highlights key contacts with Asp 189, Lys 192, and the S2 pocket.


Subject(s)
Benzoates/chemistry , Benzoates/pharmacology , Factor VIIa/antagonists & inhibitors , Factor VIIa/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/pharmacology , Thrombosis/prevention & control , Animals , Benzoates/chemical synthesis , Disease Models, Animal , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Rabbits , Structure-Activity Relationship
15.
Bioorg Med Chem Lett ; 23(18): 5244-8, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23845220

ABSTRACT

Aminoisoquinoline and isoquinoline groups have successfully replaced the more basic P1 benzamidine group of an acylsulfonamide factor VIIa inhibitor. Inhibitory activity was optimized by the identification of additional hydrophobic and hydrophilic P' binding interactions. The molecular details of these interactions were elucidated by X-ray crystallography and molecular modeling. We also show that decreasing the basicity of the P1 group results in improved oral bioavailability in this chemotype.


Subject(s)
Benzamidines , Factor VIIa/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Factor VIIa/metabolism , Humans , Models, Molecular , Molecular Structure , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship , Sulfonamides/chemical synthesis
16.
Bioorg Med Chem Lett ; 23(6): 1604-7, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23416003

ABSTRACT

Bicyclic pyrazinone and pyrimidinone amides were designed and synthesized as potent TF-FVIIa inhibitors. SAR demonstrated that the S2 and S3 pockets of FVIIa prefer to bind small, lipophilic groups. An X-ray crystal structure of optimized compound 9b bound in the active site of FVIIa showed that the bicyclic scaffold provides 5 hydrogen bonding interactions in addition to projecting groups for interactions within the S1, S2 and S3 pockets. Compound 9b showed excellent FVIIa potency, good selectivity against FIXa, Xa, XIa and chymotrypsin, and good clotting activity.


Subject(s)
Amides/chemistry , Amidines/chemical synthesis , Drug Design , Factor VIIa/antagonists & inhibitors , Pyrazines/chemistry , Pyrazines/chemical synthesis , Pyrimidinones/chemistry , Serine Proteinase Inhibitors/chemical synthesis , Amides/chemical synthesis , Amides/metabolism , Amidines/chemistry , Amidines/metabolism , Binding Sites , Bridged Bicyclo Compounds/chemistry , Catalytic Domain , Crystallography, X-Ray , Factor VIIa/metabolism , Protein Binding , Pyrazines/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism , Structure-Activity Relationship
17.
Biochem J ; 443(1): 259-66, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22239091

ABSTRACT

TFPI (tissue factor pathway inhibitor) is an anticoagulant protein that prevents intravascular coagulation through inhibition of fXa (Factor Xa) and the TF (tissue factor)-fVIIa (Factor VIIa) complex. Localization of TFPI within caveolae enhances its anticoagulant activity. To define further how caveolae contribute to TFPI anticoagulant activity, CHO (Chinese-hamster ovary) cells were co-transfected with TF and membrane-associated TFPI targeted to either caveolae [TFPI-GPI (TFPI-glycosylphosphatidylinositol anchor chimaera)] or to bulk plasma membrane [TFPI-TM (TFPI-transmembrane anchor chimaera)]. Stable clones had equal expression of surface TF and TFPI. TX-114 cellular lysis confirmed localization of TFPI-GPI to detergent-insoluble membrane fractions, whereas TFPI-TM localized to the aqueous phase. TFPI-GPI and TFPI-TM were equally effective direct inhibitors of fXa in amidolytic assays. However, TFPI-GPI was a significantly better inhibitor of TF-fVIIa than TFPI-TM, as measured in both amidolytic and plasma-clotting assays. Disrupting caveolae by removing membrane cholesterol from EA.hy926 cells, which make TFPIα, CHO cells transfected with TFPIß and HUVECs (human umbilical vein endothelial cells) did not affect their fXa inhibition, but significantly decreased their inhibition of TF-fVIIa. These studies confirm and quantify the enhanced anticoagulant activity of TFPI localized within caveolae, demonstrate that caveolae enhance the inhibitory activity of both TFPI isoforms and define the effect of caveolae as specifically enhancing the anti-TF activity of TFPI.


Subject(s)
Caveolae/metabolism , Factor VIIa/metabolism , GPI-Linked Proteins/metabolism , Lipoproteins/metabolism , Animals , Blood Coagulation , Blood Coagulation Tests , CHO Cells , Cell Membrane/metabolism , Cholesterol/deficiency , Cholesterol/metabolism , Cricetinae , Factor VIIa/antagonists & inhibitors , Factor Xa/metabolism , Factor Xa Inhibitors , Human Umbilical Vein Endothelial Cells , Humans , Protein Transport , Recombinant Fusion Proteins/metabolism , beta-Cyclodextrins
18.
Pharmazie ; 68(5): 349-54, 2013 May.
Article in English | MEDLINE | ID: mdl-23802432

ABSTRACT

AS1924269-00 is a promising orally applicable anticoagulant that inhibits FVIIa but has very low oral absorption. Therefore, in this study, we aimed to develop a prodrug of AS1924269-00, which possesses a carbamate-added amidine functional group, with high membrane permeability. We investigated the pharmacokinetics of the carbamate-type prodrug of AS1924269-00 in rats. The Caco-2 cell monolayer was used as an in vitro model and in parallel an artificial membrane permeability assay (PAMPA) was performed to examine the transport mechanisms of the prodrug. The bioavailability of the active form was determined to be only 0.3% in rats, but the oral absorption of the prodrug was markedly improved, and its bioavailability was 36%. Our in vivo result was consistent with the finding that compared to AS1924269-00, the prodrug showed favorable permeability in Caco-2 cells and PAMPA. We introduced carbamate into the amidine functional group of the FVIIa inhibitor, which possesses the amidine backbone, and converted it to a prodrug using carboxylic acid ethyl ester. This novel prodrug had favorable absorption and membrane permeability in vivo and in vitro. Thus, we suggest a clinical application of the carbamate-added amidine prodrug of the FVIIa inhibitor.


Subject(s)
Amidines/pharmacokinetics , Anticoagulants/pharmacokinetics , Factor VIIa/antagonists & inhibitors , Phenoxyacetates/pharmacokinetics , Amidines/administration & dosage , Animals , Anticoagulants/administration & dosage , Caco-2 Cells , Chromatography, High Pressure Liquid , Half-Life , Humans , Indicators and Reagents , Male , Mass Spectrometry , Membranes, Artificial , Permeability , Phenoxyacetates/administration & dosage , Rats , Rats, Sprague-Dawley
19.
Pharmazie ; 68(6): 406-13, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23875246

ABSTRACT

We recently developed a prodrug (AS1932804-00, CMP) of the novel FVIIa inhibitor AS1924269-00, which possesses a carbamate amidine backbone. In addition, we developed another type of prodrug (AS1927819-00, OXP) with an oxime amidine backbone. In this study, we investigated the efficiency of conversion of these novel FVIIa prodrugs to their active forms by evaluating the production of the active form in vitro by using microsomes, mitochondria, and cryopreserved hepatocytes, and compared it with the in vivo conversion mechanisms of the prodrugs (oxime amidine vs. carbamate amidine). We observed that OXP and CMP showed improved oral absorption, and the efficiency of conversion of CMP to the active form was higher than that of OXP. The in vivo rate of conversion of OXP to its active form was low in rats, and compared to liver microsomes and mitochondria, cryopreserved hepatocytes supplemented with serum and coenzymes were an appropriate metabolic test tool. On the other hand, the efficiency of conversion of CMP to its active from could be appropriately evaluated using small intestinal microsomes. The development of a prodrug can be optimized when information about the stability of carboxylic acid esters in the presence of serum esterases, membrane permeability of intermediate forms, and differential tissue specificity to metabolic activities for carbamate and oxime backbones of amidine can be obtained.


Subject(s)
Anticoagulants/pharmacokinetics , Factor VIIa/antagonists & inhibitors , Phenoxyacetates/pharmacokinetics , Animals , Azetidines/pharmacokinetics , Benzylamines/pharmacokinetics , Biotransformation , Hepatocytes/metabolism , In Vitro Techniques , Intestinal Mucosa/metabolism , Kinetics , Microsomes, Liver/metabolism , Mitochondria, Liver/metabolism , NAD/metabolism , NADP/metabolism , Prodrugs/metabolism , Rats
20.
Crit Care Med ; 40(4): 1229-36, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22080642

ABSTRACT

OBJECTIVE: Excessive activation of coagulation, which can culminate in overt disseminated intravascular coagulation, is a prominent feature of heat stroke. However, neither the mechanism that initiates the coagulation activation nor its pathogenic role is known. We examined whether the tissue factor/factor VIIa complex initiates the coagulation activation in heat stroke and, if so, whether upstream inhibition of coagulation activation through its neutralization may minimize cellular injury and organ dysfunction. We also examined whether coagulation inhibition influences heat stroke-induced fibrinolytic and inflammatory responses. DESIGN: Randomized controlled study. SETTING: Comparative Medicine Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia. SUBJECTS: Baboons (Papio Hamadryas). INTERVENTIONS: Twelve anesthetized baboons assigned randomly to recombinant nematode anticoagulant protein c2, a powerful inhibitor of tissue factor/factor VIIa-dependent coagulation (n = 6), or a control group (n = 6) were heat-stressed in a prewarmed neonatal incubator at 44-47°C until systolic blood pressure fell <90 mm Hg, signaling the onset of severe heat stroke. Recombinant nematode anticoagulant protein c2 was administered as a single intravenous dose of 30 µg/kg body weight at onset of heat stroke. The control group received an equivalent volume of sterile saline intravenously. MEASUREMENTS AND MAIN RESULTS: Heat stroke was associated with coagulation activation and fibrin formation as evidenced by the increased plasma thrombin-antithrombin complexes, endogenous thrombin potential, and D-dimer levels. Recombinant nematode anticoagulant protein c2 induced significant inhibition of thrombin generation and fibrin formation. Inhibition of coagulation in recombinant nematode anticoagulant protein c2-treated animals did not influence either fibrinolysis (assessed by tissue plasminogen activator, plasmin-α2-antiplasmin complexes, and plasminogen activator inhibitor) or the release of pro- and anti-inflammatory cytokines. No difference in markers of cell injury and organ dysfunction was observed between recombinant nematode anticoagulant protein c2-treated and control groups. CONCLUSIONS: Tissue factor/factor VIIa-dependent pathway initiates coagulation activation in induced-heat stroke in the baboon without an effect on fibrinolysis and inflammation. The findings suggest also that coagulation activation is not a prerequisite of cell injury and organ dysfunction.


Subject(s)
Blood Coagulation/physiology , Factor VIIa/physiology , Heat Stroke/blood , Thromboplastin/physiology , Animals , Blood Coagulation/drug effects , Blood Gas Analysis , Factor VIIa/analysis , Factor VIIa/antagonists & inhibitors , Heat Stroke/complications , Heat Stroke/physiopathology , Helminth Proteins/pharmacology , Inflammation/etiology , Inflammation/physiopathology , Papio hamadryas , Partial Thromboplastin Time , Prothrombin Time , Recombinant Proteins/pharmacology , Thrombomodulin/blood , Thromboplastin/analysis , Thromboplastin/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL