Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 259
Filter
2.
J Thromb Haemost ; 17(12): 2035-2046, 2019 12.
Article in English | MEDLINE | ID: mdl-31423724

ABSTRACT

BACKGROUND: Activated factor VII (FVIIa) is pertinent to the initiation of blood coagulation. Proteolytic and amidolytic activity of FVIIa are greatly enhanced by its cofactor, tissue factor (TF). OBJECTIVE: We aimed to generate a single-domain antibody (sdAb) that recognizes free FVIIa rather than TF-bound FVIIa. METHODS: A llama-derived phage library was used to screen for anti-FVIIa sdAbs. RESULTS: One sdAb, KB-FVIIa-004, bound to FVIIa, but not to its precursor FVII or to homologous proteins (prothrombin, factor X, or their activated derivatives). FVIIa amidolytic activity was inhibited by KB-FVIIa-004 (Ki  = 28-45 nM) in a competitive manner. KB-FVIIa-004 also inhibited FVIIa-mediated FX activation (Ki  = 26 nM). In contrast, KB-FVIIa-004 was inefficient in prolonging the clotting time of the prothrombin time-test, which was prolonged by a maximum of 10 s at high sdAb concentrations (10 µM). Furthermore, FVIIa/TF amidolytic activity or FVIIa/TF-mediated FX activation remained unaffected up to a 50-fold to 1000-fold molar excess of KB-FVIIa-004. These data suggest that KB-FVIIa-004 loses its inhibitory activity in the presence of TF. A KB-FVIIa-004/albumin fusion-protein (004-HSA) was generated for in vivo testing. By using 004-HSA, we observed that this sdAb blocked the therapeutic capacity of FVIIa to correct bleeding in FVIII-deficient mice. DISCUSSION: This observation is compatible with the view that FVIIa functions independently of TF under these conditions. In conclusion, we have generated a sdAb that specifically blocks TF-independent activity of FVIIa. This antibody can be used to gain insight into the roles of TF-bound and TF-free FVIIa.


Subject(s)
Anticoagulants/pharmacology , Blood Coagulation/drug effects , Factor VIIa/antagonists & inhibitors , Single-Domain Antibodies/pharmacology , Thromboplastin/metabolism , Animals , Coagulants/antagonists & inhibitors , Coagulants/pharmacology , Disease Models, Animal , Factor VIII/genetics , Factor VIIa/immunology , Factor VIIa/metabolism , Factor VIIa/pharmacology , Female , Hemophilia A/blood , Hemophilia A/drug therapy , Hemophilia A/genetics , Humans , Male , Mice, Inbred C57BL , Protein Binding
3.
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
4.
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
5.
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
6.
Oncotarget ; 8(16): 25915-25927, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28033108

ABSTRACT

Cancer stem cells (CSCs) are a subpopulation of cells that can self-renew and initiate tumours. The clotting-initiating protein Tissue Factor (TF) promotes metastasis and may be overexpressed in cancer cells with increased CSC activity. We sought to determine whether TF promotes breast CSC activity in vitro using human breast cancer cell lines. TF expression was compared in anoikis-resistant (CSC-enriched) and unselected cells. In cells sorted into of TF-expressing and TF-negative (FACS), and in cells transfected to knockdown TF (siRNA) and overexpress TF (cDNA), CSC activity was compared by (i) mammosphere forming efficiency (MFE) (ii) holoclone colony formation (Hc) and (iii) ALDH1 activity. TF expression was increased in anoikis-resistant and high ALDH1-activity T47D cells compared to unselected cells. FACS sorted TF-expressing T47Ds and TF-overexpressing MCF7s had increased CSC activity compared to TF-low cells. TF siRNA cells (MDAMB231,T47D) had reduced CSC activity compared to control cells. FVIIa increased MFE and ALDH1 in a dose-dependent manner (MDAMB231, T47D). The effects of FVIIa on MFE were abrogated by TF siRNA (T47D). Breast CSCs (in vitro) demonstrate increased activity when selected for high TF expression, when induced to overexpress TF, and when stimulated (with FVIIa). Targeting the TF pathway in vivo may abrogate CSC activity.


Subject(s)
Breast Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Thromboplastin/metabolism , Aldehyde Dehydrogenase 1 Family , Anoikis/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Factor VIIa/antagonists & inhibitors , Female , Gene Expression , Gene Knockdown Techniques , Humans , Isoenzymes/metabolism , Retinal Dehydrogenase/metabolism , Thromboplastin/genetics , Tumor Cells, Cultured
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.
J Med Chem ; 59(15): 7125-37, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27455395

ABSTRACT

Inhibitors of the tissue factor (TF)/factor VIIa complex (TF-FVIIa) are promising novel anticoagulants which show excellent efficacy and minimal bleeding in preclinical models. Starting with an aminoisoquinoline P1-based macrocyclic inhibitor, optimization of the P' groups led to a series of highly potent and selective TF-FVIIa inhibitors which displayed poor permeability. Fluorination of the aminoisoquinoline reduced the basicity of the P1 group and significantly improved permeability. The resulting lead compound was highly potent, selective, and achieved good pharmacokinetics in dogs with oral dosing. Moreover, it demonstrated robust antithrombotic activity in a rabbit model of arterial thrombosis.


Subject(s)
Anticoagulants/pharmacology , Drug Discovery , Factor VIIa/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Thromboplastin/antagonists & inhibitors , Administration, Oral , Animals , Anticoagulants/administration & dosage , Anticoagulants/chemistry , Biological Availability , Dogs , Dose-Response Relationship, Drug , Factor VIIa/metabolism , Healthy Volunteers , Humans , Macrocyclic Compounds/administration & dosage , Macrocyclic Compounds/chemistry , Male , Models, Molecular , Molecular Structure , Rabbits , Structure-Activity Relationship , Thromboplastin/metabolism
9.
J Med Chem ; 59(8): 4007-18, 2016 04 28.
Article in English | MEDLINE | ID: mdl-27015008

ABSTRACT

Incorporation of a methyl group onto a macrocyclic FVIIa inhibitor improves potency 10-fold but is accompanied by atropisomerism due to restricted bond rotation in the macrocyclic structure, as demonstrated by NMR studies. We designed a conformational constraint favoring the desired atropisomer in which this methyl group interacts with the S2 pocket of FVIIa. A macrocyclic inhibitor incorporating this constraint was prepared and demonstrated by NMR to reside predominantly in the desired conformation. This modification improved potency 180-fold relative to the unsubstituted, racemic macrocycle and improved selectivity. An X-ray crystal structure of a closely related analogue in the FVIIa active site was obtained and matches the NMR and modeled conformations, confirming that this conformational constraint does indeed direct the methyl group into the S2 pocket as designed. The resulting rationally designed, conformationally stable template enables further optimization of these macrocyclic inhibitors.


Subject(s)
Factor VIIa/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Serine Proteinase Inhibitors/pharmacology , Crystallography, X-Ray , Macrocyclic Compounds/chemistry , Magnetic Resonance Spectroscopy , Molecular Structure , Serine Proteinase Inhibitors/chemistry
10.
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
11.
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.
J Med Chem ; 58(15): 6225-36, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26151189

ABSTRACT

On the basis of a crystal structure of a phenylpyrrolidine lead and subsequent molecular modeling results, we designed and synthesized a novel series of macrocyclic FVIIa inhibitors. The optimal 16-membered macrocycle was 60-fold more potent than an acyclic analog. Further potency optimization by incorporation of P1' alkyl sulfone and P2 methyl groups provided a macrocycle with TF/FVIIa Ki = 1.6 nM, excellent selectivity against a panel of seven serine proteases, and FVII-deficient prothrombin time EC2x = 1.2 µM. Discovery of this potent, selective macrocyclic scaffold opens new possibilities for the development of orally bioavailable FVIIa inhibitors.


Subject(s)
Factor VIIa/antagonists & inhibitors , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Drug Design , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Models, Molecular , Molecular Structure
13.
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
14.
J Med Chem ; 58(6): 2799-808, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25764119

ABSTRACT

A multidisciplinary, fragment-based screening approach involving protein ensemble docking and biochemical and NMR assays is described. This approach led to the discovery of several structurally diverse, neutral surrogates for cationic factor VIIa P1 groups, which are generally associated with poor pharmacokinetic (PK) properties. Among the novel factor VIIa inhibitory fragments identified were aryl halides, lactams, and heterocycles. Crystallographic structures for several bound fragments were obtained, leading to the successful design of a potent factor VIIa inhibitor with a neutral lactam P1 and improved permeability.


Subject(s)
Drug Design , Factor VIIa/antagonists & inhibitors , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Blood Coagulation/drug effects , Crystallography, X-Ray , Factor VIIa/metabolism , Halogens/chemistry , Halogens/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Humans , Lactams/metabolism , Lactams/pharmacology , Models, Molecular , Molecular Docking Simulation
15.
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
16.
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
17.
J Thromb Haemost ; 12(11): 1826-37, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25163770

ABSTRACT

BACKGROUND: TFPI is a Kunitz-type protease inhibitor that downregulates the extrinsic coagulation pathway by inhibiting factor Xa (FXa) and FVIIa. All three Kunitz domains (KD1, KD2, and KD3) and protein S are required for optimal inhibition of FXa and FVIIa. There is limited information on Kunitz domain requirements of the inhibition of TF:FVIIa-catalyzed FIX and FX activation by TFPI. AIM: To investigate the role of the Kunitz domains of TFPI and protein S in the inhibition of FX and FIX activation. METHODS: Inhibition of TF:FVIIa-catalyzed FX and FIX activation by full-length TFPI (TFPIFL ) and TFPI constructs was quantified from progress curves of FXa and FIXa generation measured with chromogenic substrates. RESULTS AND CONCLUSIONS: TFPIFL inhibited TF:FVIIa-catalyzed FIX activation with a Ki of 16.7 nmol L(-1) . Protein S reduced the Ki to 1.0 nmol L(-1) . TFPI1-150 and KD1-KD2 had 10-fold higher Ki values and were not stimulated by protein S. Single Kunitz domains were poor inhibitors of TF:FVIIa-catalyzed FIX activation (Ki >800 nm). FX activation was measured at limiting FVIIa and excess TF or vice versa. At both conditions, TFPIFL , TFPI1-150 , and KD1-KD2 showed similar inhibition of FX activation. However, at low phospholipid concentrations, TFPIFL was ~ 15-fold more active than TFPI1-150 or KD1-KD2. Apparently, excess phospholipids act as a kind of sink for TFPIFL , limiting its availability for TF:FVIIa inhibition. Preformed FXa:TFPIFL/1-150 complexes rapidly and stoichiometrically inhibited FIX and FX activation by TF:FVIIa, indicating that binary TFPI:FXa complex formation is the limiting step in TF:FVIIa inhibition. Protein S also enhanced inhibition of TF:FVIIa-catalyzed FX activation by TFPI.


Subject(s)
Blood Coagulation , Factor IXa/metabolism , Factor VIIa/metabolism , Factor Xa Inhibitors/metabolism , Factor Xa/metabolism , Lipoproteins/metabolism , Thromboplastin/metabolism , Blood Coagulation/drug effects , Catalysis , Dose-Response Relationship, Drug , Factor VIIa/antagonists & inhibitors , Factor Xa Inhibitors/pharmacology , Humans , Kinetics , Lipoproteins/pharmacology , Phospholipids/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein S/metabolism , Recombinant Proteins/metabolism
18.
Drug Discov Today ; 19(9): 1440-4, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24881783

ABSTRACT

The tissue factor-coagulation factor VIIa complex (TF-fVIIa) is a well-validated biological target and has been the focus of extensive research directed toward the discovery of novel oral antithrombotics. This review briefly summarizes the key antithrombotic target validation data and provides an update on recent advances in small molecule TF-fVIIa inhibitors.


Subject(s)
Factor VIIa/antagonists & inhibitors , Fibrinolytic Agents/pharmacology , Thromboplastin/antagonists & inhibitors , Administration, Oral , Animals , Drug Design , Fibrinolytic Agents/administration & dosage , Humans
19.
Expert Opin Drug Discov ; 9(8): 859-72, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24882057

ABSTRACT

INTRODUCTION: Heparin and warfarin have historically been the only antithrombotics available. Recently, however, newer anticoagulants have been developed. Factor VIIa (fVIIa) inhibitors represent one of the new and potentially exciting classes of anticoagulants currently under development. Indeed, several methodologies have been used to develop fVIIa inhibitors. AREAS COVERED: The authors highlight some of the methologies applied for the discovery of fVIIa inhibitors including phage display, isolation of endogenous peptides from hematophagous animals and the use of the 1,5-benzothiazepine molecular scaffolds and screens of large chemical libraries previously used to identify other serine protease inhibitors. Although these screens were intended to identify thrombin and factor Xa inhibitors, the compounds often had concomitant fVIIa activity. The authors also discuss the utilization of medical chemistry techniques for the discovery of these compounds. EXPERT OPINION: FVIIa inhibitors represent a viable option for the development of new anticoagulants. There are theoretical advantages that fVIIa inhibitors may possess over existing anticoagulants and highly specific inhibitors that possess oral bioavailability and low bleeding risk may succeed.


Subject(s)
Anticoagulants/pharmacology , Drug Design , Factor VIIa/antagonists & inhibitors , Animals , Anticoagulants/adverse effects , Anticoagulants/chemistry , Biological Availability , Hemorrhage/chemically induced , Humans , Thiazepines/chemistry , Thiazepines/pharmacology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...