In-vitro evaluation of anti-factor IXa aptamer on thrombin generation, clotting time, and viscoelastometry

2009 ◽  
Vol 101 (05) ◽  
pp. 827-833 ◽  
Author(s):  
Kenichi Tanaka ◽  
Fania Szlam ◽  
Christopher Rusconi ◽  
Jerrold Levy

SummaryThe REG1 system consists of factor IXa inhibitor, RB006, an ap-tamer-based anticoagulant and its antidote, RB007. The optimal use of RB006 can be facilitated by understanding its effect on the formation of thrombin and fibrin, and other standard tests of coagulation. Blood from consented volunteers was drawn into 3.2% citrate (9:1 v/v) and either used immediately or centrifuged to obtain platelet-poor plasma. Increasing concentrations of ap-tamer (6–24 μg/ml) alone or in combination with heparin (0.1 U/ml) or lepirudin (0.2 μg/ml) were added to blood and plasma samples. Activated clotting times (ACT+, low range-ACT), thrombelastometry (ROTEM™) or thrombelastography (TEG®) were performed in recalcified whole blood samples. Thrombin generation, prothrombin time (PT) and activated partial thromboplastin time (aPTT) were performed in plasma samples. To some samples the antidote RB007 was added to neutralise the anticoagulation activity of RB006. In all experiment the ratio of RB006 to RB007 was kept 1:2. RB006 dose-dependently prolonged aPTT and low range-ACT, but, as expected, had no effect on PT. RB006 prolonged the lag time and decreased the peak of Actin-triggered thrombin generation. Thrombin-activated TEG demonstrated that RB006 decreases the rate of clot formation. These effects were potentiated when RB006 was combined with heparin or lepirudin. In all experiments RB007 reversed the effects of RB006 back to baseline. In conclusion, RB006 inhibits thrombin generation and clot formation in a concentration-dependent manner. It is feasible to monitor RB006 and its reversal with RB007 using aPTT, low range-ACT, and thrombin-activated TEG.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1877-1877
Author(s):  
Kathleen M. Welch ◽  
Kamlai Saiya-Cork ◽  
Weston R. Gould ◽  
Robert J. Leadley

Abstract Inhibition of either coagulation factor Xa (FXa) or thrombin (FIIa) alters ex vivo biomarkers of coagulation and decreases thrombus size in animal models of experimental thrombosis. The objective of this study was to determine if combining FXa and FIIa inhibitors would synergistically reduce the magnitude of IIa generated without elevating markers of bleeding. A synergistic combination may translate into lower doses, more effective anticoagulation and better safety. To predict the FXa and FIIa inhibitor combinations with the maximum potential for synergy, PD 0313052, a potent, selective FXa inhibitor, and argatroban, a potent FIIa inhibitor, were each tested independently and in combination using an in vitro thrombin generation assay. Individually, PD 0313052 and argatroban reduced total thrombin generation (TG) in a concentration dependent manner with IC50’s of 497±148 and 882±193 nM, respectively. Subsequently, PD 0313052 and argatroban were combined in 96 well plates at concentrations ranging from 0.125x to 8x their respective IC50 concentrations. Analysis using the Bliss Independence Model identified statistically significant synergistic activity, with the greatest increase (33%) over simple additivity at 249 and 441 nM FXa to FIIa, respectively, both below their respective IC50 concentrations. Furthermore, combinations of PD 0313052 and argatroban were evaluated in an assay measuring activated clotting time (ACT). Although both PD 0313052 and argatroban dose-dependently elongated the ACT, the combination of 0.5x the TG IC50 concentrations, which demonstrated the greatest synergy in the TG assay, showed the smallest increase in the ACT, prolonging clotting time by only 15%. These data demonstrate that the combination of a specific factor Xa inhibitor and a specific IIa inhibitor can synergistically reduce thrombin generation without appreciable elevation of ACT, suggesting that dual inhibition of FXa and FIIa, using relatively low doses of each, may provide efficacious and safe treatment for thromboembolic diseases.


2017 ◽  
Vol 8 (4) ◽  
pp. 525
Author(s):  
Chandra Kishore Tyagi ◽  
Deenanath Jhade ◽  
Sunil Kumar Shah

<p>The study evaluated anticoagulant properties of the aqueous extract of <em>Cestrum nocturnum</em> using aPTT-Activated Partial Thromboplastin Time, PT- Prothrombin Time &amp; TT-Thrombin Time as standard procedures.</p><p>For <em>in vitro</em> coagulation assays, aqueous extract of plant prolonged APTT, TT, and PT clotting times in a dose-dependent manner (Table 7). It prolonged APTT clotting time from 45 ± 2 (2mg/mL) to 82.2 ± 2.63s (10mg/mL), PT clotting time from 20.4 ± 1.49 (2mg/mL) to 31.4 ± 2.15s (10mg/mL), and TT clotting time from 9.2 ± 1.16 (2mg/mL) to 17.4 ± 1.01s (10mg/mL) at the concentration of 2 to 10mg/mL. Heparin prolonged APTT and PT clotting times more than 111.8s and 40.8s, respectively, at a concentration of 1 IU/mL. Heparin prolonged TT clotting times more than 20.6s at a concentration of 1 IU/mL.</p><p>The phytochemical screening of the plant confirm the presence of saponin in the water and ethanolic extract, Alkaloid in all the extract except hexane extract, tannin in water, ethanol and methanol extract, amino acid in water and ethanolic extract, carbohydrate in water and methanolic extract and triterpenoids and glycoside were absent in all the extracts. The results demonstrated that the aqueous extract of <em>Cestrum nocturnum</em> possesses pharmacologically active anticoagulant principles that could be isolated and evaluated for clinical or physiological purposes.</p>


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4151-4151
Author(s):  
Ismail Elalamy ◽  
Anna D. Petropoulou ◽  
Mohamed Hatmi ◽  
Meyer M. Samama ◽  
Grigoris T. Gerotziafas

Abstract Introduction: Orgaran® (Org 10172) is a low molecular weight heparinoid which consists of natural sulphated glycosaminoglycans (heparan, dermatan, chondroitin sulphate). It has a mean molecular weight of approximately 6 kDa (4–10 kDa), an excellent bioavailability following subcutaneous administration and an anti-Xa/anti-IIa activity ratio superior to 22. It is the anticoagulant of choice in patients developping Heparin-Induced Thrombocytopenia (HIT), whereas its’ use is also proposed for surgical thromboprophylaxis. Orgaran® has no effect on routine coagulation tests (aPTT, PT, TT). Thrombin generation test(TG) is a global clotting assay proven to be sensitive to the anticoagulant effect of LMWHs and specific FXa inhibitors (i.e. fondaparinux and BAY-597939). In this in vitro study, we determined the tissue factor (TF)-induced TG inhibition potency of Orgaran® using the Thrombogram-Thrombinoscope® assay. Materials and Methods: TG was assessed after TF pathway activation in Platelet Rich Plasma (PRP) (1.5x105 platelets/μl) using diluted thromboplastin (Dade Innovin®, 1:1000 final dilution). The clotting process is provoked by a physiologically relevant TF concentration. Orgaran® was added to control plasma from 8 healthy volunteers at five different final concentrations (0.2, 0.4, 0.6, 0.8 and 1IU anti-Xa/ml). TG was initiated by adding the triggering solution containing CaCl2 and the fluorogenic substrate. The analyzed TG parameters are the lag time, the maximal concentration of thrombin (Cmax), the time to reach Cmax (Tmax), the TG velocity and the endogenous thrombin potential (ETP). Results: Orgaran® prolonged significantly the lag time and the Tmax at a concentration over 0.40 IU anti-Xa/ml (p<0.05). At the lowest studied concentration (0.20 IU anti-Xa/ml), lag time and Tmax were only prolonged by 12%, whereas their maximal prolongation (around 50%) was observed at 1IU anti-Xa/ml. Furthermore, Orgaran® inhibited ETP, Cmax and TG velocity in an almost linear dose dependent manner. A significant inhibition of ETP, Cmax and TG velocity was obtained at concentrations superior to 0.20 IU anti-Xa/ml. (p<0.05). At the highest studied concentration (1IU anti-Xa/ml) Orgaran® suppressed all TG parameters by about 80% (Table 1). Conclusion: Orgaran® exhibited a significant inhibitory activity of in vitro TG. At concentrations achieved in clinical practice (prophylactic or therapeutic dose), Orgaran® modified in vitro TG profile while it has no effect on routine coagulation tests. Thus, TG assay is a sensitive method for monitoring Orgaran® and this test requires a clinical prospective evaluation. Table 1. Determination of IC20 and IC50 anti-Xa inhibitory concentrations of Orgaran® on TG parameters Lag Time Tmax ETP Cmax Velocity IC: Inhibitory Concentration * or Concentration increasing 20% and 50% the lag time and the Tmax respectively IC 20 (IU/ml) 0.30 0.30 0.18 0.18 0.15 IC 50 (IU/ml) 0.83 >1 0.30 0.50 0.35 1IU anti-Xa/ml 53% 47% 68% 76% 84%


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 876-876
Author(s):  
Alex Gatt ◽  
Joost van Veen ◽  
Peter Cooper ◽  
Steve Kitchen ◽  
Michael Makris

Abstract Antidotes to the ever-growing number of anticoagulants are always desirable in order to placate bleeding in emergencies. In general, the older anticoagulants like coumarins and unfractionated heparin (UFH) have proven reversal agents, whereas the newer ones do not. We studied the in vitro effect of 5 different heparinoids (UFH, Tinzaparin, Enoxaparin, Fondaparinux and Danaparoid) on the calibrated automated thrombogram. The assay uses a fluorogenic substrate that is cleaved by the thrombin formed after the addition of plasma to 5pM tissue factor, 4μM phospholipids and calcium chloride. We investigated all the parameters generated by dedicated software (Thrombinoscope™) ie the lag time (LT), the time to peak (ttpeak), the endogenous thrombin potential (ETP) and the peak thrombin. All the five anticoagulants tested inhibited thrombin generation (TG) in a concentration dependent manner. We subsequently analysed the in vitro effect of different concentrations of six potential reversal agents on correcting TG parameters of maximally inhibited plasma for each anticoagulant. These were protamine sulphate at 2.5, 5 & 8μg/ml, activated FVIIa (Novoseven®) at 5, 10 & 50μg/ml, FEIBA® at 0.5,1 & 2U/ml, Beriplex® at 0.3, 0.6 & 1.2U/ml, Prothromplex® TIM4 at 0.4, 0.8 & 1.8U/ml and fresh frozen plasma (FFP) at 250, 500 & 750μl/ml. The three concentrations reflect the recommended therapeutic doses for each agent together with lower and higher doses than normally used. As predicted, UFH (final concentration 0.527U/ml) was completely reversed with a standard protamine concentration of 5μg/ml. However, the highest dose of protamine gave slightly lower TG, indicating that higher concentrations of protamine sulphate can have a paradoxical ‘anticoagulant’ effect. High doses of FEIBA (2U/ml) and FVIIa (50μg/ml) restored ~50% of thrombin generation parameters. Tinzaparin (at 1antiXaUnit/ml) was also completely neutralised by protamine. However, a higher concentration of 8μg/ml protamine was required. This effect was not seen with Enoxaparin with this higher concentration of protamine reversing only ~40% of the ETP, 21% of the peak thrombin, 71% of ttpeak and 72% of the LT. There was no positive effect of protamine on Fondaparinux (3μg/ml) and Danaparoid (1antiXa U/ml)-treated plasma. Whereas Danaparoid seemed relatively resistant to all six reversal agents, Fondaparinux effect was completely neutralised by FVIIa at concentrations between 10–50μg/ml. This study highlights the differences in neutralisation of different low molecular weight heparins and UFH. In particular, Tinzaparin was much more readily reversed with protamine sulphate than Enoxaparin. It also indicates that high doses of FVIIa could completely reverse Fondaparinux anticoagulation.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4359-4359
Author(s):  
Gerald Schrenk ◽  
Katalin Varadi ◽  
Herbert Gritsch ◽  
Hanspeter Rottensteiner ◽  
Hartmut J. Ehrlich ◽  
...  

Abstract Abstract 4359 Baxter and Nektar have developed BAX 855, a longer-acting PEGylated form of Baxter’s recombinant FVIII (ADVATE process) using stable PEG technology from Nektar. BAX 855 was functionally characterized in vitro and its features were compared with those of the unmodified parent rFVIII. The overall hemostatic potency of BAX 855 was assessed using a thrombin generation assay. Human FVIII-deficient plasma containing less than 1% of FVIII was supplemented with different concentrations of BAX 855 and unmodified rFVIII and coagulation was triggered by adding a small amount of recombinant human tissue factor complexed with phospholipid (PL) micelles to the plasma. Similar to unmodified rFVIII, BAX 855 corrected the impaired thrombin generation of the FVIII deficient plasma in a concentration-dependent manner. The role of FVIII within the tenase complex was determined by measuring the kinetics of FXa generation with a FIXa-cofactor activity assay, using either untreated or thrombin activated BAX 855. Comparison of the kinetic parameters and the maximum FXa generated revealed similar characteristics between BAX 855 and unmodified rFVIII. A similar approach revealed that BAX 855 fully retained its ability to be activated and inactivated by thrombin. The susceptibility of BAX 855 to activated protein C (APC) inactivation was also similar for BAX 855 and unmodified rFVIII. The binding affinities for VWF were similar for unmodified rFVIII (KD 0.6 nM) and BAX 855 (KD 0.8 nM) and the binding capacity of BAX 855 was also only slightly reduced. In contrast, the binding capacity of BAX 855 to the low-density lipoprotein-receptor-related protein (LRP) clearance receptor was 55% less than that of the unmodified rFVIII. In summary, the functional properties of BAX 855 were fully retained, indicating that PEGylation did not have an impact on the functional properties of rFVIII. Disclosures: Schrenk: Baxter Innovations GmbH: Employment. Varadi:Baxter Innovations GmbH: Employment. Gritsch:Baxter Innovations GmbH: Employment. Rottensteiner:Baxter Innovations GmbH: Employment. Ehrlich:Baxter Innovations GmbH: Employment. Scheiflinger:Baxter Innovations GmbH: Employment. Turecek:Baxter Innovations GmbH: Employment.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2985-2985
Author(s):  
Neil A. Goldenberg ◽  
William E. Hathaway ◽  
Linda Jacobson ◽  
Marilyn J. Manco-Johnson

Abstract BACKGROUND: The Clot Formation and Lysis (CloFAL) assay, modified from prior global assay methods by He et al., 1999, and Smith et al., 2003, involves activation of coagulation and fibrinolysis in platelet-poor plasma via addition of a physiologic reactant solution in a multi-well microassay plate. Continuous spectropohotometric data analysis allows measurement of kinetic absorbance changes of the plasma sample over three hours, which yields a unique clot formation and lysis curve. Using parameters of the curve, coagulation and fibrinolytic indices (CI and FI) are calculated relative to a simultaneously run pooled normal plasma standard. METHODS: Platelet-poor plasma obtained from pregnant women at term (n=24), neonatal cord blood (n=29), and healthy children (n=22) were analyzed using the CloFAL assay. Healthy adult (n=22) plasma samples, as well as those of individuals with factor deficiencies, were obtained commercially. Fibrinolytic alterations in vitro were also investigated. Intra-assay coefficients of variation (CVs) for normal controls ranged from 3–12% for all assay parameters, with inter-assay CVs of 5–15%. RESULTS: Representative CloFAL curves for healthy adults and children, pregnant women, and newborn infants are shown in Figure 1. Coagulation potential (measured by median CI) was significantly increased, while fibrinolytic capacity (measured by median FI) was markedly decreased, in pregnant women as compared to healthy adults (CI: 239% vs. 115%, FI: 59% vs. 95%; P&lt;0.001 for each). By contrast, CI was decreased, and FI notably increased, in neonatal cords versus children, although the former comparison did not achieve statistical significance (CI: 58% vs. 69%, P=0.09; FI: 210% vs. 142%, P&lt;0.001). The influence of deficiencies of coagulation factors and fibrinolytic regulators upon CloFAL parameters was also investigated. The greatest impact upon CI occurred with severe deficiency of fibrinogen or factors II, V, VII, VIII, IX, or X. Furthermore, CI was sensitive to deficiencies of factor VIII and fibrinogen in a concentration-dependent manner. In addition, FI was increased by PAI-1 deficiency and by inhibition of thrombin-activatable fibrinolysis inhibitor (TAFI) activation, and was zero in the setting of aminocaproic acid treatment. Further studies revealed that the influence of heparin concentrations of up to 2 U/mL in plasma was completely reversible by heparinase treatment of samples prior to assay. CONCLUSION: These results indicate that the CLoFAL assay is reproducible and analytically sensitive to known physiologic and pathologic alterations in coagulation and fibrinolysis. The application of this global assay to patients with a variety of disorders of thrombosis and hemostasis is currently ongoing. Figure Figure


2008 ◽  
Vol 412 (3) ◽  
pp. 545-551 ◽  
Author(s):  
Jorge G. Ganopolsky ◽  
Sophie Charbonneau ◽  
Henry T. Peng ◽  
Pang N. Shek ◽  
Mark D. Blostein

On the basis of previous evidence that amphipathic helical peptides accelerate Factor IXa activation of Factor X [Blostein, Rigby, Furie, Furie and Gilbert (2000) Biochemistry 39, 12000–12006], the present study was designed to assess the procoagulant activity of an IAP (ideal amphipathic peptide) of Lys7Leu15 composition. The results show that IAP accelerates Factor X activation by Factor IXa in a concentration-dependent manner and accelerates thrombin generation by Factor Xa with a comparable peptide- and substrate-concentration-dependence. A scrambled helical peptide with the same amino acid composition as IAP, but with its amphipathicity abolished, eliminated most of the aforementioned effects. The Gla (γ-carboxyglutamic acid)-rich domain of Factor X is required for IAP activity, suggesting that this peptide behaves as a phospholipid membrane. This hypothesis was confirmed, using fluorescence spectroscopy, by demonstrating direct binding between IAP and the Gla-rich domain of Factor X. In addition, the catalytic efficiencies of the tenase and prothrombinase enzymatic complexes, containing cofactors Factor VIIIa and Factor Va respectively, are enhanced by IAP. Finally, we show that IAP delays clot lysis in vitro. In summary, these observations demonstrate that IAP not only enhances essential procoagulant reactions required for fibrin generation, but also inhibits fibrinolysis, suggesting a potential role for IAP as a haemostatic agent.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 528-528 ◽  
Author(s):  
Elisabeth Perzborn ◽  
Michaela Harwardt

Abstract Activation of protein C is dependent on thrombin complexed with thrombomodulin (TM). Activated protein C (APC), together with its cofactor protein S, degrades coagulation Factors Va and VIIIa, thereby limiting further thrombin formation. Thus, in addition to suppressing the procoagulant effects of thrombin, direct thrombin inhibitors (DTIs) may also downregulate anticoagulant effects of thrombin-mediated feedback mechanisms. By contrast, direct Factor Xa (FXa) inhibitors block the formation of thrombin, but not its actions. The objective of this study was to investigate whether the direct FXa inhibitor, rivaroxaban, and the DTIs, dabigatran and melagatran, inhibit the negative-feedback reaction of the thrombin–TM complex/APC (thrombin–TM/APC) system and thereby increase thrombin formation. Experiments were conducted in plateletpoor plasma from healthy donors (normal plasma) and in pooled protein C-deficient plasma, both substituted with 1.33 μM phospholipids, in the presence or absence of 10 nM TM with increasing concentrations of rivaroxaban, dabigatran, melagatran, or the appropriate vehicles. Thrombin formation was initiated by adding 1.67 pM tissue factor (TF) and assessed by measuring the cleavage of the fluorogenic substrate Z-Gly-Gly-Arg-AMC (Bachem) using the Calibrated Automated Thrombogram (CAT, Thrombinoscope® BV) method. The parameters assessed were lag time, time to peak thrombin generation (tmax), peak thrombin generation (Cmax), and endogenous thrombin potential (ETP). In addition, formation of prothrombin fragments 1+2 (F1+2) was determined by ELISA (Enzygnost® F1+2 monoclonal [Dade Behring]). Rivaroxaban potently inhibited thrombin formation in the absence and presence of TM across all parameters in a concentration-dependent manner in both normal plasma and protein C-deficient plasma (see Table). In the absence of TM, melagatran and dabigatran also inhibited thrombin formation in a concentration-dependent manner, both in normal plasma and protein C-deficient plasma. In the presence of TM, DTIs prolonged lag time and tmax in a concentration-dependent manner. However, only high concentrations of the DTIs reduced ETP, Cmax, and F1+2, In normal plasma,lower concentrations even increased ETP, Cmax, and F1+2. Increased thrombin formation was observed with melagatran 119–474 nM or dabigatran 68–545 nM. DTIs did not increase thrombin formation in protein C-deficient plasma, suggesting that both protein C and TM are needed for the DTI-mediated increase in thrombin formation. The results suggest that low concentrations of DTIs suppress the anticoagulant effects of the thrombin–TM/APC system by inhibiting activation of protein C by the thrombin–TM complex, and thereby enhance thrombin formation. Conversely, rivaroxaban does not increase thrombin formation, suggesting that it does not suppress the negative-feedback reaction by inhibition of protein C activation. This hypothesis is supported by the absence of enhanced thrombin formation in protein C-deficient plasma. Enhanced thrombin formation might explain the hypercoagulation observed with DTIs in a rat model of TF-induced intravascular coagulation (Furugohri, et al. 2005; Morishima, et al. 2005; Perzborn, et al. 2008) and suggests that DTIs could cause activation of coagulation at lower plasma concentrations. Table. Effect of rivaroxaban, dabigatran, and melagatran on peak thrombin formation (Cmax [nM thrombin]) in the absence or presence of thrombomodulin (TM) in normal plasma (NP) from healthy volunteers (n=8–12), and in pooled protein C-deficient plasma (PPC) in the presence of TM (n=3). Results were obtained by the CAT method and are a mean of n plasma samples. Prothrombin fragments F1+2 (nM F1+2) results were obtained in the presence of TM (mean results; n=3 [NP]). Thrombin (nM) in normal and in protein C-deficient plasma n.d:, no data. Rivaroxaban (nM) 0 18 91 182 363 1,090 Cmax: − TM in NP 273 222 113 71 43 15 Cmax: + TM in NP 100 67 32 18 10 3 Cmax: + TM in PPC 290 249 168 117 80 38 F1+2: + TM (in NP) 184 89 43 18 n.d. 2 Dabigatran (nM) 0 68 136 273 545 1,090 Cmax: − TM in NP 261 287 289 282 239 102 Cmax: + TM in NP 81 156 203 240 218 96 Cmax: + TM in PPC 298 301 300 292 253 116 F1+2: + TM in NP 252 n.d. 362 422 351 98 Melagatran(nM) 0 24 119 237 474 948 Cmax: − TM in NP 276 290 297 289 251 127 Cmax: + TM in NP 101 133 215 251 237 113 Cmax: + TM in PPC 294 296 299 292 256 132 F1+2: + TM in NP 213 n.d. 389 427 393 123


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 384-384 ◽  
Author(s):  
Catherine J Rea ◽  
Benny Sorensen ◽  
Jørgen Ingerslev ◽  
Peter Laursen

Abstract Abstract 384 Fibrinogen: A Procoagulant and an Anticoagulant Introduction: Bleeding occurs secondary to acquired fibrinogen deficiency but the effect of high fibrinogen is more controversial. Correlation between raised fibrinogen levels and venous or arterial thrombosis has been recorded. However, fibrinogen increases as an acute phase response and may be an innocent biomarker, detected at elevated levels in individuals with concomitant disease. Recent animal studies provide evidence that high fibrinogen does not trigger thrombosis per se, but enhances thrombotic occlusion of vessels following tissue injury. Aims: This study aims to investigate the effect of elevated levels of fibrinogen on thrombin generation and clot resistance to accelerated fibrinolysis. We hypothesised that fibrinogen promotes clot stability following a high tissue factor stimulus (TF), but will act as an anticoagulant following low TF stimulus. Method: Normal human plasma was spiked with fibrinogen to achieve final plasma concentrations of 2.7, 3.2, 3.7, 4.7, 5.7, 6.7, 8 and 11.7g/l. Coagulation was initiated with TF at variable dilutions (1:20000, 1: 500) plus calcium. To assess clot stability the same assay was performed with simultaneous addition of tissue plasminogen activator (t-Pa 0.75nmolar). Clot formation and lysis was recorded via light absorbance (FLUOstar Omega). Clot stability was also measured by whole blood thromboelastometry; citrate and CTI stabilized whole blood was drawn from a healthy individual and spiked with fibrinogen (calculated plasma concentrations: 3.2, 4, 3.9, 5.5, 9.9, 16 g/l). Coagulation was triggered with TF (1: 50000 or 1:500), calcium and of t-Pa (2nmolar). The area under elasticity curve (AUEC) at 90mins was the primary endpoint. Thrombin generation in plasma was performed in plasma following addition of fibrinogen using fluorogenic substrate and calcium (FluCä, Thrombinoscope BV, The Netherlands). Results:Plasma clot formation assay: Dose dependent shortening of clot time and time to peak turbidity were seen with increasing fibrinogen following a high TF stimulus (TF 1:500) (figure 1- panel A). Conversely, following a low TF stimulus increasing fibrinogen caused a lengthening of the clotting time. Plasma and whole blood lysis assays: With high TF stimulus, fibrinogen produced a dose-dependent increase in clot stability measures (AUC/AUEC) in both plasma and whole blood assays (figure 1-panel B). Following a low TF stimulus increases in the fibrinogen concentration resulted in suppressed clot stability. Thrombin generation: A decrease in total thrombin generation was seen with increasing fibrinogen (Figure 2) at both high and low TF levels. With high TF there was no alteration in lag-time, but with low TF stimulus the lag-time progressively lengthened as fibrinogen concentration increased. Discussion: Fibrinogen acts as a pro-coagulant by promoting clot formation and supports clot stability following a high TF stimulus. However, following a low TF stimulus elevated fibrinogen becomes an anticoagulant as demonstrated by prolonging clotting time and decreases clot stability in both plasma and whole blood. In conclusion, our data suggest that elevated fibrinogen per se is not thrombogenic. However, following a significant trauma resulting in a high tissue factor stimulus and high thrombin generation, fibrinogen acts predominantly as a pro-coagulant enhancing clot formation and supporting clot stability. This may protect against bleeding or contribute to pathological thrombotic events. In contrast, following a minor trauma prompting a minimal tissue factor stimulus, fibrinogen predominantly acts as an anticoagulant and may protect against thrombosis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2002 ◽  
Vol 99 (3) ◽  
pp. 923-930 ◽  
Author(s):  
Saulius Butenas ◽  
Kathleen E. Brummel ◽  
Richard F. Branda ◽  
Sara G. Paradis ◽  
Kenneth G. Mann

Abstract The ability of factor VIIa to initiate thrombin generation and clot formation in blood from healthy donors, blood from patients with hemophilia A, and in anti–factor IX antibody–induced (“acquired”) hemophilia B blood was investigated. In normal blood, both factor VIIa–tissue factor (TF) complex and factor VIIa alone initiated thrombin generation. The efficiency of factor VIIa was about 0.0001 that of the factor VIIa–TF complex. In congenital hemophilia A blood and “acquired” hemophilia B blood in vitro, addition of 10 to 50 nM factor VIIa (pharmacologic concentrations) corrected the clotting time at all TF concentrations tested (0-100 pM) but had little effect on thrombin generation. Fibrinopeptide release and insoluble clot formation were only marginally influenced by addition of factor VIIa. TF alone had a more pronounced effect on thrombin generation; an increase in TF from 0 to 100 pM increased the maximum thrombin level in “acquired” hemophilia B blood from 120 to 480 nM. Platelet activation was considerably enhanced by addition of factor VIIa to both hemophilia A blood and “acquired” hemophilia B blood. Thus, pharmacologic concentrations of factor VIIa cannot restore normal thrombin generation in hemophilia A and hemophilia B blood in vitro. The efficacy of factor VIIa (10-50 nM) in hemophilia blood is dependent on TF.


Sign in / Sign up

Export Citation Format

Share Document