Platelet-dependent thrombin generation assay for monitoring the efficacy of recombinant Factor VIIa

Platelets ◽  
2005 ◽  
Vol 16 (1) ◽  
pp. 45-50 ◽  
Author(s):  
W. Wegert ◽  
S. Harder ◽  
S. Bassus ◽  
C. M. Kirchmaier
Blood ◽  
2008 ◽  
Vol 112 (8) ◽  
pp. 3227-3233 ◽  
Author(s):  
Cees Weeterings ◽  
Philip G. de Groot ◽  
Jelle Adelmeijer ◽  
Ton Lisman

Abstract Several lines of evidence suggest that recombinant factor VIIa (rFVIIa) is able to activate factor X on an activated platelet, in a tissue factor-independent manner. We hypothesized that, besides the anionic surface, a receptor on the activated platelet surface is involved in this process. Here, we showed that, in an ELISA setup, a purified extracellular fragment of GPIbα bound to immobilized rFVIIa. Surface plasmon resonance established a affinity constant (Kd) of approximately 20 nM for this interaction. In addition, CHO cells transfected with the GPIb-IX-V complex could adhere to immobilized rFVIIa, whereas wild-type CHO cells could not. Furthermore, platelets sti-mulated with a combination of collagen and thrombin adhered to immobilized rFVIIa under static conditions. Platelet adhesion was inhibited by treatment with O-sialoglycoprotein endopeptidase, which specifically cleaves GPIbα from the platelet surface. In addition, rFVIIa-mediated thrombin generation on the activated platelet surface was inhibited by cleaving GPIbα from its surface. In summary, 3 lines of evidence showed that rFVIIa interacts with the GPIb-IX-V complex, and this interaction enhanced tissue factor-independent thrombin generation mediated by rFVIIa on the activated platelet surface. The rFVIIa-GPIbα interaction could contribute to cessation of bleeding after administration of rFVIIa to patients with bleeding disorders.


Blood ◽  
2003 ◽  
Vol 101 (5) ◽  
pp. 1864-1870 ◽  
Author(s):  
Ton Lisman ◽  
Sultana Moschatsis ◽  
Jelle Adelmeijer ◽  
H. Karel Nieuwenhuis ◽  
Philip G. De Groot

A novel approach to treat bleeding episodes in patients with Glanzmann thrombasthenia (GT) and perhaps also in patients receiving αIIbβ3 inhibitors is the administration of recombinant factor VIIa (rFVIIa). The mechanism of action of rFVIIa in these patients is, however, still unclear. We studied the effect of rFVIIa-mediated thrombin formation on adhesion of αIIbβ3-deficient platelets under flow conditions. Adhesion of αIIbβ3-deficient platelets to the extracellular matrix (ECM) of stimulated human umbilical vein endothelial cells or to collagen type III was studied using a model system with washed platelets and red cells. When αIIbβ3-deficient platelets were perfused over the surface at arterial shear rate for 5 minutes, a low surface coverage was observed (GT platelets, mean ± SEM, 37.5% ± 5.0%; normal platelets preincubated with an RGD-containing peptide, 7.4% ± 2.1%). When rFVIIa, together with factors X and II, was added to the perfusate, platelet deposition significantly increased (GT platelets, mean ± SEM, 67.0% ± 4.3%; normal platelets preincubated with an RGD-containing peptide, 48.2% ± 2.9%). The same effect was observed when normal platelets were pretreated with the commercially available anti-αIIbβ3 drugs abciximab, eptifibatide, or tirofiban. It was shown that tissue factor–independent thrombin generation (presumably induced by binding of rFVIIa to adhered platelets) was responsible for the increase in platelet deposition. In conclusion, defective adhesion of αIIbβ3-deficient platelets to ECM can be restored by tissue factor–independent rFVIIa-mediated thrombin formation. The enhanced generation of platelet procoagulant surface facilitates fibrin formation, so that lack of platelet aggregate formation might be compensated for.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2618-2618
Author(s):  
Ton Lisman ◽  
Jelle Adelmeijer ◽  
Sandra Cauwenberghs ◽  
Elisabeth C.M. van Pampus ◽  
Johan W.M. Heemskerk ◽  
...  

Abstract Recombinant factor VIIa (rFVIIa) has been developed for treatment of inhibitor-complicated hemophilia. Increasing clinical evidence shows that rFVIIa also appears safe and effective as pro-hemostatic agent in various other clinical disorders including patients with thrombocytopenia. Relatively little is known about the mechanism of action of rFVIIa in hemophilia and its various other potential indications. It is generally accepted that rFVIIa functions by enhancement of thrombin generation at the site of injury. It is, however, unknown if and how this affects platelet adhesion and aggregation. Previously, we have shown that rFVIIa-mediated thrombin generation substantially enhances deposition of platelets from patients with Glanzmann thrombasthenia (a qualitative or quantitative deficiency of the integrin αIIbβ3) to subendothelial proteins (Blood2004, 103: 1720). Here, we investigated the effect of rFVIIa-mediated thrombin generation on platelet adhesion and aggregation under flow conditions at normal and reduced platelet count. A mixture of washed platelet and red cells was perfused over collagen or fibrinogen in presence or absence of a thrombin-generating system consisting of purified coagulation factors rFVIIa, X, and prothrombin in the presence of calcium chloride. Addition of this thrombin-generating system enhanced platelet adhesion and aggregation to collagen and adhesion and spreading to fibrinogen at normal platelet count and at platelet numbers as low as 10.000/μl. rFVIIa-mediated thrombin generation enhanced the activation state of platelets adhered under flow as measured by intracellular calcium fluxes, and enhanced the exposure of procoagulant phospholipids as measured by annexin A5 binding. It was shown that FITC-labeled rFVIIa bound to collagen-adhered platelets under flow conditions in experiments in which whole blood from severely thrombocytopenic patients was used. We believe thrombin is generated by platelet-bound rFVIIa independent of tissue factor. Taken together, increased platelet adhesion and aggregation by rFVIIa-mediated thrombin formation may explain the therapeutic effects of rFVIIa in thrombocytopenic conditions and in patients with a normal platelet count by (1) enhancement of primary hemostasis and (2) enhancement of procoagulant surface leading to elevated fibrin formation. These platelet-promoting effects of rFVIIa might also, in part, explain the therapeutic effects of rFVIIa in other indications.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1207-1207
Author(s):  
Sarah T.B.G. Loubele ◽  
Henri M.H. Spronk ◽  
Rene van Oerle ◽  
Hugo ten Cate ◽  
Peter L.A. Giesen

Abstract Abstract 1207 Background: Although the recombinant factor VIIa (rFVIIa) has been registered for use in hemophilia patients with inhibitors, there is still no method to monitor the effects of rFVIIa in restoring the coagulation balance in plasma. Hence, information is lacking about the individual optimal dose needed to normalize thrombin generation. Methods: The calibrated automated thrombogram (CAT) method was modified to increase sensitivity for rFVIIa addition to plasma at concentrations of 0, 2.5, 5, 10, 20, 40 and 80 nM, which covers the expected plasma concentration of 26 nM reached after standard administration. Thrombin generation was triggered using combinations of TF concentrations between 0 and 4 pM, and phospholipids concentrations between 0 and 4 μM. Endogenous thrombin potential (ETP), peak height, and velocity index were calculated in platelet poor plasmas (PPP) of different donors. All blood was collected in citrated tubes containing corn trypsin inhibitor (CTI) to minimize any contact activation. Results: The optimal conditions for discriminating rFVIIa (0–80 nM) in the CAT assay were determined in PPP: 0 or 0.25 pM TF with 4 μM of phospholipids. Also at higher TF concentrations, the CAT method was able to detect varying rFVIIa concentrations. The optimal concentration of phospholipids was 4 μM for all TF concentrations. In plasma of 6 healthy volunteers, thrombin generation triggered with 0.25 pM TF dose dependently increased using varying rFVIIa concentrations between 0 and 80 nM (Figure 1, left panel). The mean values for ETP, peak height and velocity index are depicted in Table 1. On average, addition of 2.5, 5, 10, 20, 40 or 80 nM of rFVIIa resulted in a 146, 156, 161, 174, 206, and 285 % of the peak height compared to 0 nM rFVIIa, which was set at 100 %. At 4 pM TF the maximum ETP, peak height, and velocity index were reached at concentrations less than 20 nM rFVIIa for all donors. The mean values are depicted in Table 2. Surprisingly, at 80 nM rFVIIa, thrombin generation was decreased compared to lower rFVIIa concentrations (Figure 1, right panel). Addition of 2.5, 5, 10, 20, 40 or 80 nM of rFVIIa resulted in 107, 109, 107, 103, 100, or 94 % of the peak height without addition of rFVIIa (0 nM set at 100 %). In FVIII deficient patient plasma (PPP), this effect was also present and even more pronounced. Here, a dose dependent effect of rFVIIa addition was visible at low (0 or 0.25 pM) TF trigger, whereas at 4 pM TF trigger ETP, peak height and velocity index were maximal in the presence of 10 nM rFVIIa. Overall, the peak height was 136, 142, 126, 102, 94, and 81 % upon addition of 5, 10, 20, 30, 40, or 80 nM rFVIIa respectively compared to 0 nM rFVIIa (set to 100 %). Discussion: In hemophilic as well as normal plasma, the addition of rFVIIa dose dependently altered thrombin generation triggered with a low TF trigger (0 or 0.25 pM). At a higher trigger of 4 pM TF, maximal thrombin generation was obtained at rFVIIa concentrations of less then 20 nM. Remarkably, thrombin generation was attenuated in the presence of 80 nM rFVIIa. This paradox may be explained by assuming that the endogenously activated VIIa is more active than the rVIIa that was added. At higher rVIIa dosages the fraction of TF occupied by endogenous VIIa will decrease resulting in less active TF:VIIa complexes. This effect will be more pronounced when FXa formation is dependent on TF:FVIIa alone without the involvement of the tenase complex, which shows from our analysis in hemophilic plasma. Overall, these data suggest that the assay is most sensitive to added rVIIa when the contribution to Xa formation of TF:VIIa complex is small compared to that of rVIIa alone, i.e., in conditions where there is no or very little TF present. Disclosures: Giesen: Thrombinoscope bv: Employment.


2004 ◽  
Vol 91 (06) ◽  
pp. 1090-1096 ◽  
Author(s):  
Marcel Levi ◽  
Troy Sarich ◽  
Stig Boström ◽  
Ulf Eriksson ◽  
Maria Eriksson-Lepkowska ◽  
...  

SummaryThe objectives were to investigate whether activation of the extrinsic coagulation cascade by recombinant factor VIIa (rFVIIa) reverses the inhibition of thrombin generation and platelet activation by melagatran, the active form of the oral direct thrombin inhibitor ximelagatran. In a single-blind, randomized, parallel-group study, volunteers (20 per group) received a 5-hour intravenous (iv) infusion to achieve steadystate melagatran plasma concentrations of approximately 0.5 µmol/L, with a single iv bolus of rFVIIa (90 µg/kg) or placebo at 60 minutes. Prothrombin fragment 1+2, thrombin-antithrombin complex, fibrinopeptide A, β-thromboglobulin, and thrombin-activatable fibrinolysis inhibitor were quantified for venous and shed blood. Activated partial thromboplastin time (APTT), prothrombin time (PT), endogenous thrombin potential, thrombus precursor protein (TpP), and plasmin-α2-antiplasmin complex concentrations were determined in venous blood. Shed blood volume was measured. Melagatran reduced markers of thrombin generation and platelet activation in shed blood and prolonged APTT. rFVIIa increased FVIIa activity, PT, and TpP in venous blood. All other parameters were unaffected. In conclusion, rFVIIa did not reverse the anticoagulant effects of high constant concentrations of melagatran. However, the potential value of higher, continuous or repeated doses of rFVIIa or its use with lower melagatran concentrations has not been excluded.


Sign in / Sign up

Export Citation Format

Share Document