Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
J Thromb Haemost ; 16(6): 1099-1106, 2018 06.
Article in English | MEDLINE | ID: mdl-29575637

ABSTRACT

Essentials Statins lower venous thromboembolism risk in general but have not been studied in cancer patients. We completed a randomized trial of rosuvastatin vs. placebo among cancer patients on chemotherapy. Rosuvastatin did not significantly lower prothrombotic biomarkers including D-dimer. The role of statins in venous thrombosis prevention in cancer patients remains unknown. SUMMARY: Background Statin therapy is associated with lower risk of venous thromboembolism (VTE) but has not been prospectively evaluated in patients with advanced cancer. Objectives We determined if statin administration in this high-risk population reduces the risk of VTE, based on established and emerging biomarkers. Patients/Methods This double-blind, crossover, randomized controlled trial among patients with advanced cancer receiving systemic therapy allocated participants to rosuvastatin 20 mg daily or placebo for 3-4 weeks prior to crossover to the alternative therapy, with a 3-5-week washout. D-dimer, C-reactive protein (CRP), soluble (s)P-selectin, factor VIII (FVIII), thrombin generation and exploratory biomarkers focusing on endogenous thrombin potential, including tissue factor (TF), activated factor IX (FIXa) and activated factor XI (FXIa), were measured at the start and end of both treatment periods. The primary outcome was change in D-dimer with rosuvastatin compared with placebo. Results Of 38 enrolled participants, 24 (63%) completed the study. Rosuvastatin did not cause statistically significant changes in D-dimer levels or any other biomarker. CRP levels decreased by 40%; 4.3 mg L-1 (95% confidence interval, -11.0 to +2.5 mg L-1 ) compared with placebo. In post-hoc analysis, participants who received rosuvastatin initially during their first line of treatment had a 13% decrease in D-dimer. Circulating TF, FIXa and FXIa were detected in 26%, 68% and 71% of cancer patients despite not being found in healthy individuals. Conclusions Rosuvastatin did not cause favorable changes in biomarkers of VTE risk in advanced cancer patients receiving chemotherapy. The role of statin therapy as thromboprophylaxis in the cancer population remains uncertain.


Subject(s)
Antineoplastic Agents/therapeutic use , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinolytic Agents/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Neoplasms/drug therapy , Rosuvastatin Calcium/therapeutic use , Venous Thromboembolism/prevention & control , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Biomarkers/blood , C-Reactive Protein/metabolism , Cross-Over Studies , Double-Blind Method , Factor IXa/metabolism , Factor VIII/metabolism , Factor XIa/metabolism , Female , Fibrinolytic Agents/adverse effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Male , Middle Aged , Neoplasms/blood , Neoplasms/complications , Neoplasms/diagnosis , P-Selectin/blood , Risk Factors , Rosuvastatin Calcium/adverse effects , Thrombin/metabolism , Thromboplastin/metabolism , Time Factors , Treatment Outcome , Venous Thromboembolism/blood , Venous Thromboembolism/diagnosis , Venous Thromboembolism/etiology , Vermont
2.
J Thromb Haemost ; 14(10): 2001-2010, 2016 10.
Article in English | MEDLINE | ID: mdl-27431334

ABSTRACT

Essentials Acidosis, an outcome of traumatic injury, has been linked to impaired procoagulant efficiency. In vitro model systems were used to assess coagulation dynamics at pH 7.4 and 7.0. Clot formation dynamics are slightly enhanced at pH 7.0 in blood ex vivo. Acidosis induced decreases in antithrombin efficacy offset impairments in procoagulant activity. SUMMARY: Background Disruption of hydrogen ion homeostasis is a consequence of traumatic injury often associated with clinical coagulopathy. Mechanisms by which acidification of the blood leads to aberrant coagulation require further elucidation. Objective To examine the effects of acidified conditions on coagulation dynamics using in vitro models of increasing complexity. Methods Coagulation dynamics were assessed at pH 7.4 and 7.0 as follows: (i) tissue factor (TF)-initiated coagulation proteome mixtures (±factor [F]XI, ±fibrinogen/FXIII), with reaction progress monitored as thrombin generation or fibrin formation; (ii) enzyme/inhibitor reactions; and (iii) TF-dependent or independent clot dynamics in contact pathway-inhibited blood via viscoelastometry. Results Rate constants for antithrombin inhibition of FXa and thrombin were reduced by ~ 25-30% at pH 7.0. At pH 7.0 (+FXI), TF-initiated thrombin generation showed a 20% increase in maximum thrombin levels and diminished thrombin clearance rates. Viscoelastic analyses showed a 25% increase in clot time and a 25% reduction in maximum clot firmness (MCF). A similar MCF reduction was observed at pH 7.0 when fibrinogen/FXIII were reacted with thrombin. In contrast, in contact pathway-inhibited blood (n = 6) at pH 7.0, MCF values were elevated 6% (95% confidence interval [CI]: 1%-11%) in TF-initiated blood and 15% (95% CI: 1%- 29%) in the absence of TF. Clot times at pH 7.0 decreased 32% (95% CI: 15%-49%) in TF-initiated blood and 51% (95% CI: 35%-68%) in the absence of TF. Conclusions Despite reported decreased procoagulant catalysis at pH 7.0, clot formation dynamics are slightly enhanced in blood ex vivo and suppression of thrombin generation is not observed. A decrease in antithrombin reactivity is one potential mechanism contributing to these outcomes.


Subject(s)
Acidosis/blood , Blood Coagulation Tests/methods , Blood Coagulation/drug effects , Thrombin/pharmacology , Antithrombin III/analysis , Blood Coagulation Disorders , Elasticity , Fibrin/analysis , Fibrinogen/pharmacology , Healthy Volunteers , Humans , Hydrogen-Ion Concentration , Ions , Proteome , Thrombin/antagonists & inhibitors , Thromboplastin/pharmacology , Time Factors , Viscosity
3.
Haemophilia ; 22(3): 462-73, 2016 May.
Article in English | MEDLINE | ID: mdl-26822998

ABSTRACT

INTRODUCTION: Factor VIII (FVIII) products used in haemophilia A treatment show inter-and intra-product and inter-assay differences in specific activity. The mechanistic basis of these differences remains unclear. AIM: The aim of this study was to mechanistically compare the functional properties of an in-house excipient-free full-length FVIII standard and pharmacologic recombinant products containing full-length (products A and B) or B-domainless (C and D) FVIII. METHODS: Factor VIII protein concentration was quantitated by ELISA. Product potency determinations (APTT, intrinsic tenase assays) and kinetic analyses detailing these products' activations by thrombin and FXa, their spontaneous and activated protein C (APC) catalysed inactivation and their performances in coagulation proteome reconstructions were studied +/- von Willebrand factor (VWF). Computational models were developed to facilitate interpretation of empirical data. RESULTS: Factor VIII protein content per manufacturer activity unit was highest for product C with the other three products similar to the standard. Potency estimates, done five different ways, varied 20-30% in inter- and intra-assay comparisons, with product B consistently showing lower specific activity. Kinetic analyses showed the five FVIII species to differ somewhat in maximum rate of activation, the maximum level of activity achieved, the rate of spontaneous or APC catalysed inactivation and the magnitude of the effect of VWF on these parameters. When evaluated both computationally and empirically in the context of tissue factor initiated thrombin generation, product C appears the most dissimilar. CONCLUSION: Assessments of FVIII activation/inactivation dynamics report larger differences between FVIII products than standard functional assays. However, all FVIII products promote a 'normal' thrombin generation response to TF.


Subject(s)
Factor VIII/metabolism , Catalysis , Coagulants/therapeutic use , Enzyme-Linked Immunosorbent Assay , Factor VIII/genetics , Factor VIII/therapeutic use , Factor Xa/metabolism , Hemophilia A/drug therapy , Humans , Kinetics , Partial Thromboplastin Time , Protein C/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/therapeutic use , Thrombin/metabolism , von Willebrand Factor/metabolism
4.
Haemophilia ; 22(2): 240-247, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26517283

ABSTRACT

BACKGROUND: Inhibitor formation complicates haemophilia treatment and requires immune tolerance induction to rid inhibitors over 5 BU. In the prospective, randomized International Immune Tolerance Study, immune tolerance induction was equally effective with high-dose (HD) (200 IU kg-1 day-1 ) and low-dose (LD) (50 IU kg-1 3× per week) factor VIII, but haemorrhages were twofold higher in the LD arm. This finding was unexpected as inhibitors neutralize FVIII activity. We hypothesized that the thrombin generation assay (TGA), a global measure of clot formation, might predict bleeding better than FVIII levels. METHODS: We evaluated TGA using relipidated tissue factor (TF) on 83 thawed, recalcified corn trypsin inhibitor/citrate plasma samples from 31 subjects (17 HD, 14 LD) who participated on the ITI study, and who had sufficient sample available and appropriate informed consent. RESULTS: There were no significant differences in peak thrombin, estimated thrombin potential, maximum rate or lag time between HD and LD arms; between pre-, during and post-ITI time points, or after FVIII spiking. In 19 subjects (12 HD, 7 LD) with anti-FVIII<1.0 BU, the prevalence of non-neutralizing antibody (NNA) and neutralizing antibody (NA) was 89.5% (17/19), and the latter strongly correlated with anti-VIII titer, r = 0.73 [95% CI: 0.55, 0.88]. CONCLUSION: In haemophilia inhibitor patients, thrombin generation is present, but does not predict bleeding risk. Following tolerance induction, NNA remains detectable in the majority.

5.
J Thromb Haemost ; 13(10): 1867-77, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26286125

ABSTRACT

BACKGROUND: The relationships of thrombin generation (TG) with cardiovascular disease risk are underevaluated in population-based cohorts. OBJECTIVES: To evaluate the relationships of TG influenced by the contact and tissue factor coagulation pathways ex vivo with common single-nucleotide polymorphisms (SNPs) and incident cardiovascular disease and stroke. PATIENTS/METHODS: We measured peak TG (pTG) in baseline plasma samples of Cardiovascular Health Study participants (n = 5411), both with and without inhibitory anti-factor XIa antibody (pTG/FXIa(-) ). We evaluated their associations with ~ 50 000 SNPs by using the IBCv2 genotyping array, and with incident cardiovascular disease and stroke events over a median follow-up of 13.2 years. RESULTS: The minor allele for an SNP in the FXII gene (F12), rs1801020, was associated with lower pTG in European-Americans (ß = - 34.2 ± 3.5 nm; P = 3.3 × 10(-22) ; minor allele frequency [MAF] = 0.23) and African-Americans (ß = - 31.1 ± 7.9 nm; P = 9.0 × 10(-5) ; MAF = 0.42). Lower FXIa-independent pTG (pTG/FXIa(-) ) was associated with the F12 rs1801020 minor allele, and higher pTG/FXIa(-) was associated with the ABO SNP rs657152 minor allele (ß = 16.3 nm; P = 4.3 × 10(-9) ; MAF = 0.37). The risk factor-adjusted ischemic stroke hazard ratios were 1.09 (95% confidence interval CI 1.01-1.17; P = 0.03) for pTG, 1.06 (95% CI 0.98-1.15; P = 0.17) for pTG/FXIa(-) , and 1.11 (95% CI 1.02-1.21; P = 0.02) for FXIa-dependent pTG (pTG/FXIa(+) ), per one standard deviation increment (n = 834 ischemic strokes). In a multicohort candidate gene analysis, rs1801020 was not associated with incident ischemic stroke (ß = - 0.02; standard error = 0.08; P = 0.81). CONCLUSIONS: These results support the importance of contact activation pathway-dependent TG as a risk factor for ischemic stroke, and indicate the importance of F12 SNPs for TG ex vivo and in vivo.


Subject(s)
Blood Coagulation/genetics , Brain Ischemia/genetics , Factor XII/genetics , Stroke/genetics , Thrombin/metabolism , Black or African American/genetics , Age Factors , Aged , Brain Ischemia/blood , Brain Ischemia/ethnology , Factor XII/metabolism , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Incidence , Male , Phenotype , Polymorphism, Single Nucleotide , Prospective Studies , Risk Assessment , Risk Factors , Stroke/blood , Stroke/ethnology , Time Factors , United States/epidemiology , White People/genetics
7.
Haemophilia ; 19(4): 619-25, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23557464

ABSTRACT

The development of anti-factor (F)VIII antibodies in haemophilia A (HA) subjects undergoing replacement therapy has been well documented. The correlation between antibody development and the FVIII product used for replacement therapy remains a subject of discussion. The aim of this study was to evaluate the presence of anti-FVIII antibodies towards three commercial rFVIII products in 34 HA subjects' plasmas. Antibodies were quantitated by a Multiplex Fluorescence Immunoassay. All plasmas contained anti-FVIII antibodies at variable concentrations ranging from 50 nm to 570 µm. Eleven of the 20 HA subjects treated with one (r)FVIII product contained inhibitory anti-FVIII antibodies (0.8-3584 BU). The inhibitory antibody titre and the molar concentrations of total antibody were mildly correlated (r(2) = 0.6). Pronounced differences in antibody recognition with the three rFVIII products were observed. For the group treated with Product 'A', the titre towards this product was 2.4-fold higher than that observed with another full-length rFVIII-containing product (Product 'B') and almost four-fold higher than that measured with a B domain-less rFVIII product (Product 'C'). For the group of 14 HA subjects treated with FVIII other than Product 'A', only one showed higher antibody titre when measured with this product. Our data suggest that the development of anti-FVIII antibodies is biased towards the product used for treatment and that a significant fraction of antibodies bind to the B domain of FVIII.


Subject(s)
Antibodies/blood , Factor VIII/immunology , Adolescent , Adult , Child , Child, Preschool , Factor VIII/antagonists & inhibitors , Factor VIII/therapeutic use , Hemophilia A/blood , Hemophilia A/drug therapy , Hemophilia A/immunology , Humans , Infant , Male , Middle Aged , Recombinant Proteins/therapeutic use , Young Adult
8.
Anal Biochem ; 422(1): 46-51, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22266209

ABSTRACT

Thrombelastography (TEG) is a method that is used to conduct global assays that monitor fibrin formation and fibrinolysis and platelet aggregation in whole blood. The purpose of this study was to use a well-characterized tissue factor (Tf) reagent and contact pathway inhibitor (corn trypsin inhibitor, CTI) to develop a reproducible thrombelastography assay. In this study, blood was collected from 5 male subjects (three times). Clot formation was initiated in whole blood with 5 pM Tf in the presence of CTI, and fibrinolysis was induced by adding tissue plasminogen activator (tPA). Changes in viscoelasticity were then monitored by TEG. In quality control assays, our Tf reagent, when used at 5 pM, induced coagulation in whole blood in 3.93 ± 0.23 min and in plasma in 5.12 ± 0.23 min (n=3). In TEG assays, tPA significantly decreased clot strength (maximum amplitude, MA) in all individuals but had no effect on clot time (R time). The intraassay variability (CVa<10%) for R time, angle, and MA suggests that these parameters reliably describe the dynamics of fibrin formation and degradation in whole blood. Our Tf reagent reproducibly induces coagulation, making it an ideal tool to quantify the processes that contribute to mechanical clot strength in whole blood.


Subject(s)
Blood Coagulation Tests/methods , Blood Coagulation , Elasticity , Thrombelastography/methods , Adult , Blood Cell Count , Blood Viscosity , Fibrin/chemistry , Fibrinolysis , Humans , Male , Plant Proteins/chemistry , Platelet Activation , Quality Control , Reproducibility of Results , Thromboplastin/chemistry , Time Factors
9.
J Thromb Haemost ; 9(11): 2262-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21920012

ABSTRACT

BACKGROUND: Replacement therapy for hemophilic patient treatment is costly, because of the high price of pharmacologic products, and is not affordable for the majority of patients in developing countries. OBJECTIVE: To generate and evaluate low molecular weight agents that could be useful for hemophilia treatment. METHODS: Potential agents were generated by synthesizing specific inhibitors [6-(Lys-Lys-Thr-[homo]Arg)amino-2-(Lys[carbobenzoxy]-Lys[carbobenzoxy]-O-benzyl)naphthalenesulfonamide] (PNASN-1)] for activated protein C (APC) and tested in plasma and fresh blood from hemophilia A patients. RESULTS: In the activated partial thromboplastin time-based APC resistance assay, PNASN-1 partially neutralized the effect of APC. In calibrated automated thrombography, PNASN-1 neutralized the effect of APC on thrombin generation in normal and congenital factor VIII-deficient plasma (FVIII:C < 1%). The addition of PNASN-1 to tissue factor-triggered (5 pm) contact pathway-inhibited fresh blood from 15 hemophilia A patients with various degrees of FVIII deficiency (FVIII:C < 1-51%) increased the maximum level of thrombin generated from 78 to 162 nm, which approached that observed in blood from a healthy individual (201 nm). PNASN-1 also caused a 47% increase in clot weight in hemophilia A blood. CONCLUSIONS: Specific APC inhibitors compensate to a significant extent for FVIII deficiency, and could be used for hemophilia treatment.


Subject(s)
Hemophilia A/drug therapy , Protein C Inhibitor/pharmacology , Thrombin/biosynthesis , Blood/drug effects , Blood Coagulation/drug effects , Blood Coagulation Tests , Case-Control Studies , Drug Evaluation , Humans , Partial Thromboplastin Time , Protein C Inhibitor/chemical synthesis , Thrombin/drug effects
10.
Haemophilia ; 17(4): 636-40, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21299745

ABSTRACT

Up to one-third of haemophilia A patients develop factor VIII (FVIII) alloantibodies (inhibitors). The Bethesda assay detects inhibitors but is relatively insensitive. Recently, a new fluorescence-based immunoassay (FLI) was developed for antibody detection. The aim of this study was to assess the prevalence of inhibitors as measured by FLI. Assays of FVIII, FVIII inhibitor by Bethesda assay with Nijmegen modification, and FVIII inhibitor by FLI were performed on adult patients with haemophilia A. Data were complete for 46 patients (median age 39), of whom 72% were severe, 7% moderate and 22% mild. The Bethesda assay was positive in only two patients (4%), while FLI was positive in 23 of 46 patients (50%), with values ranging from 0.4 to 33.7 nm (median 3.5 nm). FLI titres exceeded 7.0 nm in 19.5% of patients, all but one of whom had severe haemophilia. FLI antibody-positive patients were less likely to be HIV positive (30% vs. 70%, P = 0.02). The use of a prophylaxis regimen was associated with a lower incidence of antibody; only two of 23 patients with detectable antibody and none of those with antibody >7 nm were on a prophylaxis regimen, while nine of 23 patients without antibody were on prophylaxis, (P = 0.03). There was no difference in inhibitor presence in patients using recombinant versus plasma-derived factor. Antibodies detected by FLI are frequent in patients with haemophilia A, but are less common in those who are HIV positive or are receiving regular FVIII prophylaxis.


Subject(s)
Factor VIII/immunology , Hemophilia A/immunology , Isoantibodies/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Female , Fluorescent Antibody Technique/methods , HIV Seropositivity/immunology , Humans , Immunoassay/methods , Isoantibodies/analysis , Male , Middle Aged , Young Adult
11.
J Thromb Haemost ; 8(8): 1745-53, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20492473

ABSTRACT

BACKGROUND: Therapeutic agents that regulate blood coagulation are critical to the management of thrombotic disorders, with the selective targeting of factor (F) Xa emerging as a promising approach. OBJECTIVE: To assess anticoagulant strategies targeting FXa. METHODS: A deterministic computational model of tissue factor (Tf)-initiated thrombin generation and two empirical experimental systems (a synthetic coagulation proteome reconstruction using purified proteins and a whole blood model) were used to evaluate clinically relevant examples of the two available types of FXa-directed anticoagulants [an antithrombin (AT)-dependent agent, fondaparinux, and an AT-independent inhibitor, Rivaroxaban] in experimental regimens relevant to long-term (suppression of new Tf-initiated events) and acute (suppression of ongoing coagulation processes) clinical applications. RESULTS: Computational representations of each anticoagulant's efficacy in suppressing thrombin generation over a range of anticoagulant concentrations in both anticoagulation regimens were validated by results from corresponding empirical reconstructions and were consistent with those recommended for long-term and acute clinical applications, respectively. All three model systems suggested that Rivaroxaban would prove more effective in the suppression of an ongoing coagulation process than fondaparinux, reflecting its much higher reactivity toward the prothrombinase complex. CONCLUSION: The success of fondaparinux in acute settings in vivo is not explained solely by its properties as an FXa inhibitor. We have reported that FIXa contributes to the long-term capacity of clot-associated catalysts to restart a coagulation process, suggesting that the enhanced anti-FIXa activity of fondaparinux-AT may be critical to its success in acute settings in vivo.


Subject(s)
Anticoagulants/therapeutic use , Blood Coagulation/drug effects , Factor Xa Inhibitors , Computer Simulation , Factor V/chemistry , Factor Xa/chemistry , Fondaparinux , Humans , Morpholines/therapeutic use , Polysaccharides/therapeutic use , Recombinant Proteins/therapeutic use , Rivaroxaban , Software , Thiophenes/therapeutic use , Thrombin/biosynthesis , Time Factors , Treatment Outcome
13.
J Thromb Haemost ; 7 Suppl 1: 181-6, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19630796

ABSTRACT

We have developed an integrated approach that combines empirical and computational methodologies to define an individual's thrombin phenotype. We have evaluated the process of thrombin generation in healthy individuals and individuals with defined pathologies in order to develop general criteria relevant to assess an individual's propensity for hemorrhage or thrombosis. Three complementary hypotheses have emerged from our work: (i) compensation by the ensemble of other coagulation proteins in individuals with specific factor deficiencies can 'normalize' an individual's thrombin generation process and represents a rationale for their unexpected phenotype; (ii) individuals with clinically unremarkable factor levels may present thrombin generation profiles typical of individuals with hemostatic complications; and (iii) in some hemostatic disorders a specific pattern of expression of a small ensemble of coagulation factors may be sufficient to explain the overall phenotype.


Subject(s)
Empirical Research , Models, Theoretical , Thrombin/physiology , Computational Biology , Hemorrhage , Hemostasis , Humans , Phenotype , Thrombin/biosynthesis , Thrombosis
14.
J Thromb Haemost ; 7(5): 825-32, 2009 May.
Article in English | MEDLINE | ID: mdl-19192107

ABSTRACT

BACKGROUND: The conversion of fibrinogen to fibrin and its crosslinking to form a stable clot are key events in providing effective hemostasis. OBJECTIVES: To evaluate the relationship of fibrinopeptide (FP) release and factor (F) XIII activation in whole blood from hemophiliacs. PATIENTS/METHODS: We investigated FPA and FPB release, FXIII activation and fibrin mass in tissue factor-initiated coagulation in whole blood from individuals with hemophilia and healthy subjects. RESULTS: In hemophiliacs, the rates of fibrin formation were delayed as compared to healthy individuals. FPA/FPB release and FXIII activation were decreased in hemophiliacs vs. healthy individuals: 5.4 +/- 0.7 microM min(-1) to 1.7 +/- 0.4 microM min(-1) (P = 0.003), 2.3 +/- 0.6 microM min(-1) to 0.5 +/- 0.1 microM min(-1) (P = 0.025), and 12.1 +/- 0.7 nM min(-1) to 3.1 +/- 0.7 nM min(-1) (P < 0.0005), respectively. More FPA was released in hemophiliacs (6.6 +/- 1.2 microM) prior to clot time (CT) than in healthy individuals (2.6 +/- 0.4 microM, P = 0.013), whereas FPB and activated FXIII levels remained comparable. FXIII activation, which normally coincides with FPA release, was delayed in hemophiliacs. At CT in normal blood, the FPA concentration was 2.6-fold higher than that of FPB (P = 0.003), whereas in hemophiliacs this ratio was increased to 6.6-fold (P = 0.001). CONCLUSIONS: These data suggest that essential dynamic correlations exist between the presentations of fibrin I, fibrin II, and FXIIIa. The 'discordance' of fibrin formation in hemophiliacs results in clots that are more soluble than normal (43% lower mass; P = 0.02). The resulting poor physical clot strength probably plays a crucial role in the pathology of hemophilia.


Subject(s)
Fibrin/biosynthesis , Hemophilia A/metabolism , Adult , Case-Control Studies , Factor XII/metabolism , Humans , Male
15.
Hamostaseologie ; 29(1): 7-16, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19151839

ABSTRACT

Our studies involve computational simulations, a reconstructed plasma/platelet proteome, whole blood in vitro and blood exuding from microvascular wounds. All studies indicate that in normal haemostasis, the binding of tissue factor (TF) with plasma factor (F) VIIa (extrinsic FXase complex) results in the initiation phase of the procoagulant response. This phase is negatively regulated by tissue factor pathway inhibitor (TFPI) in combination with antithrombin (AT) and the protein C (PC) pathway. The synergy between these inhibitors provides a threshold-limited reaction in which a stimulus of sufficient magnitude must be provided for continuation of the reaction. With sufficient stimulus, the FXa produced activates some prothrombin. This initial thrombin activates the procofactors and platelets required for presentation of the intrinsic FXase (FVIIIa-FIXa) and prothrombinase (FVa-FXa) complexes which drive the subsequent propagation phase; continuous downregulation of which is provided by AT and the thrombin-thrombomodulin-PC complex. FXa generation during the propagation phase is largely (>90%) provided by the intrinsic FXase complex. TF is required for the initiation phase of the reaction but becomes non-essential once the propagation phase has been achieved. The propagation phase catalysts (FVIIIa-FIXa and FVa-FXa) continue to drive the reaction as blood is resupplied to the wound site by flow. Ultimately, the control of the reaction is governed by the pro- and anticoagulant dynamics and the supply of blood reactants to the site of a perforating injury. Our systems have been utilized to examine the qualities of hypothetical and novel antihaemorrhagic and anticoagulation agents and in epidemiologic studies of venous and arterial thrombosis and haemorrhagic pathology.


Subject(s)
Blood Coagulation/physiology , Hemostasis/physiology , Blood Coagulation Factors/metabolism , Blood Proteins/metabolism , Fibrinogen/metabolism , Humans , Kinetics , Models, Biological , Proteome , Thrombin/metabolism
16.
J Thromb Haemost ; 7(1): 132-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18983499

ABSTRACT

BACKGROUND: It has been reported by multiple laboratories that the quantitation of factor (F)VIII by activity-based assays is influenced by the method, procedure and the quality of reagents used in the assays. OBJECTIVE: To evaluate the influence of von Willebrand factor (VWF) on FVIII activity in vitro. METHODS: The activated partial thromboplastin time (APTT) and synthetic coagulation proteome assays were used. Citrated FVIII/VWF-depleted substrate plasma (SP) (both antigens < 0.5%) was used in all APTT assays. RESULTS: The concentration of FVIII antigen in pooled plasma from healthy donors [normal plasma (NP)] was 1.5 nm. The SP reconstituted with 1.5 nm recombinant (r)FVIII clotted in 23.8 +/- 0.2 s (standard deviation). The addition of 10 microg mL(-1) VWF to the SP increased the clotting time to 28.7 +/- 0.1 s; that is, the activity of rFVIII (FVIIIc) decreased to 50%. This inhibitory effect of VWF decreased with decreasing rFVIII concentration in SP, and became negligible at rFVIII

Subject(s)
Blood Coagulation/drug effects , Factor VIII/pharmacology , von Willebrand Factor/pharmacology , Blood Coagulation Tests , Dose-Response Relationship, Drug , Factor VIII/antagonists & inhibitors , Humans , Partial Thromboplastin Time
17.
Haemophilia ; 15(1): 63-72, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18691375

ABSTRACT

Several factor (F) VIII products of different origin and structure are being used for haemophilia A treatment worldwide. The assessment of FVIII concentration in these products is done using activity assays, which are dependent upon the assay and its modifications. To evaluate FVIII products for potency and for FVIII concentration and specific activity, three activity-based assays [activated partial thromboplastin time (APTT), intrinsic FXase and synthetic coagulation proteome] and two immunoassays (ELISA and western blotting) were used in this study with albumin-free full-length recombinant (r) FVIII as a standard. In all activity assays, products A and B (both contain full-length rFVIII) at 1 U mL(-1) showed potency similar to that of the 0.7 nm (1 U mL(-1)) rFVIII standard. Product E (contains truncated rFVIII) was less potent in the APTT (83% of standard) and product C (contains plasma FVIII) was less potent in FXase assays (66%). The ELISA immunoassay revealed that the specific activity of FVIII proteins in products A-C and E varied over a wide range (3900-13 200 U mg(-1)) and was higher for most lots when compared with the standard (5000 U mg(-1)), whereas the specific activity of product D (contains plasma FVIII) was lower than expected (3200-4800 U mg(-1)). (i) FVIII potency estimated in different assays gives dissimilar results; (ii) the specific activity of FVIII in various FVIII products is different and inconsistent. Thus, the administration of an equal FVIII potency in units means the administration of different amounts of FVIII protein, which may partly explain apparent discrepancies in product performance.


Subject(s)
Factor VIII/pharmacology , Blotting, Western/methods , Cysteine Endopeptidases/chemistry , Enzyme-Linked Immunosorbent Assay/methods , Factor VIII/analysis , Factor VIII/standards , Humans , Male , Neoplasm Proteins/chemistry , Partial Thromboplastin Time , Proteome , Recombinant Proteins/analysis , Recombinant Proteins/pharmacology , Reference Standards
18.
J Thromb Haemost ; 6(1): 104-10, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17944993

ABSTRACT

BACKGROUND: Acute coronary syndrome (ACS) is associated with thrombin formation, triggered by ruptured or eroded coronary atheroma. We investigated whether thrombin generation based on circulating coagulation protein levels, could distinguish between acute and stable coronary artery disease (CAD). METHODS AND RESULTS: Plasma coagulation factor (F) compositions from 28 patients with ACS were obtained after onset of chest pain. Similar data were obtained from 25 age- and sex-matched patients with stable CAD. All individuals took aspirin. Patients on anticoagulant therapy were excluded. The groups were similar in demographic characteristics, comorbidities and concomitant treatment. Using each individual's coagulation protein composition, tissue factor (TF) initiated thrombin generation was assessed both computationally and empirically. TF pathway inhibitor (TFPI), antithrombin (AT), factor II (FII) and FVIII differed significantly (P < 0.01) between the groups, with levels of FII, FVIII and TFPI higher and AT lower in ACS patients. When thrombin generation profiles from individuals in each group were compared, simulated maximum thrombin levels (P < 0.01) and rates (P < 0.01) were 50% higher with ACS while the initiation phases of thrombin generation were shorter. Empirical reconstructions of the populations reproduced the thrombin generation profiles generated by the computational model. The differences between the thrombin generation profiles for each population were primarily dependent upon the collective contribution of AT, FII and FVIII. CONCLUSION: Simulations of thrombin formation based on plasma composition can discriminate between acute and stable CAD.


Subject(s)
Acute Coronary Syndrome/blood , Blood Coagulation Factors/analysis , Coronary Artery Disease/blood , Thrombin/biosynthesis , Acute Coronary Syndrome/diagnosis , Antithrombin III , Coronary Artery Disease/diagnosis , Diagnosis, Differential , Factor VIII , Humans , Models, Statistical , Prothrombin
19.
J Thromb Haemost ; 5(10): 2055-61, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17883701

ABSTRACT

BACKGROUND: Sodium citrate has been used as an anticoagulant to stabilize blood and blood products for over 100 years, presumably by sequestering Ca(++) ions in vitro. Anticoagulation of blood without chelation can be achieved by inhibition of the contact pathway by corn trypsin inhibitor (CTI). OBJECTIVE: To evaluate the influence of citrate anticoagulation on the performance of blood, platelet-rich and platelet-poor plasma assays. METHODS: Blood was anticoagulated in three ways: by collection into citrate, CTI and citrate with CTI. Plasma was prepared using each anticoagulation regimen. Functional analyses included calibrated automated thrombography, thromboelastography, plasma clotting, the synthetic coagulation proteome and platelet aggregation. Coagulation reactions were initiated with tissue factor-phospholipid and Ca(++) (when indicated). RESULTS: In all cases, citrate anticoagulation resulted in reaction dynamics significantly altered relative to blood or plasma stabilized with CTI alone. Subsequent experiments showed that calcium citrate itself impairs coagulation dynamics. CONCLUSION: Coagulation analyses using blood that has been exposed to citrate and recalcified do not yield reliable depictions of the natural dynamics of blood coagulation processes.


Subject(s)
Anticoagulants/therapeutic use , Citrates/therapeutic use , Thrombin/chemistry , Blood Coagulation , Blood Coagulation Tests , Calcium/metabolism , Calcium Citrate/pharmacology , Calibration , Humans , Ions , Platelet Activation , Platelet Aggregation , Thrombelastography/methods , Thrombin/metabolism , Thrombin Time
20.
J Thromb Haemost ; 5(5): 1084-5; author reply 1085-7, 2007 May.
Article in English | MEDLINE | ID: mdl-17355574
SELECTION OF CITATIONS
SEARCH DETAIL
...