scholarly journals Ibrutinib treatment affects collagen and von Willebrand factor-dependent platelet functions

Blood ◽  
2014 ◽  
Vol 124 (26) ◽  
pp. 3991-3995 ◽  
Author(s):  
Marie Levade ◽  
Elodie David ◽  
Cédric Garcia ◽  
Pierre-Alexandre Laurent ◽  
Sarah Cadot ◽  
...  

Key Points Ibrutinib affects collagen and VWF-mediated platelet activation. The bleeding diathesis correlates with defects in collagen-induced platelet aggregation and firm adhesion on VWF at arterial shear rate.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3296-3296 ◽  
Author(s):  
Loic Ysebaert ◽  
Marie Levade ◽  
Garcia Cedric ◽  
Anne-Sophie Michallet ◽  
Constantine Tam ◽  
...  

Abstract Introduction Ibrutinib is the first-in-class covalent inhibitor of Bruton's Tyrosine Kinase (BTK), now approved for the therapy of mantle cell lymphoma (MCL) and chronic lymphocytic leukemia (CLL). Mild bleeding disorders (grade 1-2) have been reported in 44-60% of patients across clinical trials, with <5% grade 3 hemorrhages after trauma. After vascular injury, platelets adhere onto von Willebrand factor (vWF, through GPIb-IX-V complex) and collagen (through a2b1 and GPVI receptor), and activate phospholipase Cg2 (PLCg2) through BTK phosphorylation. In this study, we sought to examine phosphorylation pathways and platelet functions in vitro and ex vivo from ibrutinib-treated patients. Patients and Methods Within the compassionate access program of ibrutinib in France (started Feb 2014), we investigated whether ibrutinib could impact on platelet functions in vitro and ex vivo, as measured at day 0 and day 15-30 by: aggregometry using various agonists, measurement of intra-cellular levels of phosphorylation of BTK and PLCg2 phosphorylations, monitoring adhesion onto vWF matrix under high shear rate. We next assess how in vitro tests could help identify bleeding risk in a larger cohort of patients from three institutions. Results First, we demonstrated that in healthy donors' platelets, ibrutinib inhibits collagen and collagen related peptide (CRP) -induced platelet aggregation in a dose-dependent manner (mean EC50=250nM, a dose achievable in patients). This effect was paralleled by the inhibition of PLCg2 phosphorylation on the Btk-dependent phosphorylation site Tyr753, and of the auto-phosphorylation Tyr223 site of BTK itself, suggesting a specific targeting by ibrutinib. Of note, adhesion on vWF under high shear rate was dramatically decreased. In parallel, in 7/14 patients had bleeding symptoms (5/7 with grade 1-2 bleedings) and they all presented a strong inhibition of platelet aggregation in response to collagen and a significant decrease in adhesion onto vWF. Thus, the easy-to-use collagen-induced platelet aggregation test in platelet rich plasma could help physicians to decide when to perform surgical procedures without haemostasis concerns. Moreover, we show that addition of 50% untreated platelets is sufficient to efficiently reverse the effects of ibrutinib, and that platelet functions recover following treatment interruption as physiological platelet renewal occurs, supporting the in vitro data. On the other hand, patients who received aspirin (n=6) had no cases of severe bleeding and no significant impact on collagen/CRP-induced platelet aggregation. Because aspirin+P2Y12 inhibitors (such as clopidogrel, Plavix®) is widely used in the elderly population, ibrutinib therapy should be given very cautiously to these patients (who receive then three major pathway platelet activation pathway inhibitor), as recommended for vitamin K antagonists drugs. Aggregometry tests may provide important information to physicians to predict the bleeding risk as observed in our cohort of >30 patients (as of June 2014, recruitment still ongoing). Two last points should be emphasized when considering bleeding risk of ibrutinib: (i) from our study, some patients had no anti-platelet detectable effect ex vivo under ibrutinib therapy, the mechanism of which still remains unclear, and (ii) in patients with mild bleedings, platelet functions recovery and cessation of symptoms occured in virtually all patients (except those on aspirin therapy) after 3-6 months despite ongoing lymphoma responses, suggesting a potential adaptative process in platelets. Summary and Conclusion We identified that ibrutinib affects collagen and Von Willebrand Factor-mediated platelet activation in vitro and ex vivo. The mild bleeding diathesis observed in a subgroup of ibrutinib-treated patients correlates with defects in collagen-induced platelet aggregation and platelet adhesion on von Willebrand Factor at high shear rate. Based on in vitro analyses and in vivo platelet turnover, 2-3 days ibrutinib cessation appears to be enough for effective aggregation response recovery, and reintroduction of the drug should be rapid to avoid disease recurrence. Our study also suggests that platelet transfusion at a dose sufficient to get 50 % of fresh platelets may correct haemostasis in emergency, provided it was given after elimination of ibrutinib from blood (4-6h). Disclosures Tam: Pharmacyclics and Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2003 ◽  
Vol 102 (6) ◽  
pp. 2122-2129 ◽  
Author(s):  
Shuju Feng ◽  
Julio C. Reséndiz ◽  
Xin Lu ◽  
Michael H. Kroll

Abstract We examined the hypothesis that filamin A binding to the cytoplasmic tail of platelet glycoprotein Ibα (GpIbα) is regulated by pathologic shear stress and modulates von Willebrand factor (VWF)–induced platelet activation. To begin, we examined filamin binding to GpIbα in Chinese hamster ovary cells coexpressing mutant human GpIb-IX and wild-type human filamin A. We observed that many different deletions and truncations N-terminal to GpIbα's cytoplasmic domain residue 594 disrupted filamin A binding, but that binding was unaffected by 14 different point mutations in hydrophilic residues between amino acids 557 and 593. To try to narrow GpIbα's filamin A–binding domain, we next measured the effect of several cytoplasmic domain peptides on human filamin A binding to a GST-GpIbα cytoplasmic domain fusion protein. One peptide (residues 557-575; designated “A4 peptide”) inhibited filamin A binding to the GST-GpIbα cytoplasmic domain fusion protein and competed with GpIbα for binding to filamin A. When the A4 peptide was delivered to intact human platelets using a carrier peptide, we observed the dose-dependent inhibition of VWF-induced platelet aggregation in response to both ristocetin and shear stress. The effect of the A4 peptide on shear-induced platelet aggregation was accompanied by the attenuation of shear-induced filamin A binding to GpIbα and diminished shear-dependent protein tyrosine phosphorylation. These results suggest that shear-dependent VWF-induced platelet activation affects filamin A binding to GpIb-IX-V, and that filamin A binding to the cytoplasmic tail of GpIbα regulates proaggregatory tyrosine kinase signaling.


Blood ◽  
1990 ◽  
Vol 76 (2) ◽  
pp. 345-353 ◽  
Author(s):  
RR Hantgan ◽  
G Hindriks ◽  
RG Taylor ◽  
JJ Sixma ◽  
PG de Groot

We have investigated the molecular basis of thrombus formation by measuring the extent of platelet deposition from flowing whole blood onto fibrin-coated glass coverslips under well-defined shear conditions in a rectangular perfusion chamber. Platelets readily and specifically adhered to fibrin-coated coverslips in 5 minute perfusion experiments done at either low (300 s-1) or high (1,300 s-1) wall shear rates. Scanning electron microscopic examination of fibrin-coated coverslips after perfusions showed surface coverage by a monolayer of adherent, partly spread platelets. Platelet adhesion to fibrin was effectively inhibited by a monoclonal antibody (MoAb) specific for glycoprotein (GP) IIb:IIIa. The dose-response curve for inhibition of adhesion by anti-GPIIb:IIIa at both shear rates paralleled that for inhibition of platelet aggregation. Platelet aggregation and adhesion to fibrin were also blocked by low concentrations of prostacyclin. In contrast, anti- GPIb reduced adhesion by 40% at 300 s-1 and by 70% at 1,300 s-1. A similar pattern of shear rate-dependent, incomplete inhibition resulted with a MoAb specific for the GPIb-recognition region of von Willebrand factor (vWF). Platelets from an individual with severe von Willebrand's disease, whose plasma and platelets contained essentially no vWF, exhibited defective adhesion to fibrin, especially at the higher shear rate. Addition of purified vWF restored adhesion to normal values. These results are consistent with a two-site model for platelet adhesion to fibrin, in which the GPIIb:IIIa complex is the primary receptor, with GPIb:vWF providing a secondary adhesion pathway that is especially important at high wall shear rates.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3101-3101
Author(s):  
Yoshihiko Sakurai ◽  
Midori Shima ◽  
Shogo Kasuda ◽  
Shoko Omura ◽  
Masahiro Takeyama ◽  
...  

Abstract Background: The replacement therapy with plasma-derived factor VIII (FVIII)/von Willebrand factor (VWF) concentrates is the first line treatment for the patients with type 3 von Willebrand disease (VWD). However, development of anti-VWF alloantibodies (inhibitor) is a major problem since the inhibitor neutralizes the VWF activity and may cause anaphylactic reactions. As an alternative treatment, the usage of FVIII concentrates has been reported but the mechanism of the hemostatic effects remains to be elucidated. Objectives: The purpose of this study is to address the role of FVIII in the hemostatic mechanism in the absence of VWF by in vitro and ex vivo analysis in the treatment for type 3 VWD with recombinant FVIII (rFVIII). Patient/Methods: The patient is a 55-year-old male with type 3 VWD. Blood samples were obtained before and 30 min after bolus administration. Rotating thromboelastometry (ROTEM) assay was performed to examine global interactions in hemostasis. To elucidate the effect on platelet activation, α-thrombin- and shear-induced platelet aggregation studies were performed. Further, α-thrombin-induced [Ca2+]i rise was assessed using fura2-AM loaded platelets. Results and Implications: The patient underwent two surgical procedures of multiple teeth extractions successfully with minimal bleeding by bolus administration of rFVIII (50 IU/kg) before procedure and followed by continuous infusion at rate of 10 IU/kg/h for 15 hours. FVIII:C was elevated from 1.0% to 20~30% 30 min after bolus infusion and maintained ~15% after 12 h-continuous infusion. ROTEM analysis showed that infusion of rFVIII shortened clotting time (preinfusion 2083.8±784.3 sec vs. post-infusion 1022.0±191.5 sec) and clot formation time (pre 1267.3±455.4 sec vs. post 705.8±261.8 sec) and increased α (pre 8.5±7.4 degree vs. post 23.5±4.4 degree). The α value and CFT indicate the rate of increase of elastic shear modulus. Addition of rFVIII to preinfusion blood in vitro corrected ROTEM parameters and thrombin-induced aggregation dose-dependently. Infusion of FVIII enhanced thrombin-induced platelet aggregation (% maximal aggregation: pre 26.3% vs. post 98.2%) as well as low shear-induced platelet aggregation (% maximal aggregation: pre 18% vs. post 52%). Furthermore, infusion of rFVIII meliorated thrombin-induced intracellular calcium flux of washed platelets (thrombin 10 nM, Ca flux: pre 414.0 nM vs. post 620.6 nM). Recently, the cell-based model of hemostasis provides a solid foundation for the relation between platelet and coagulation. Although coagulation initiation occurs normally via the extrinsic pathway, amplification mediated by the intrinsic pathway is seriously disturbed in type 3 VWD due to the marked decrease in FVIII. Therefore, correction of FVIII could result in the improvement of hemostasis. Our data demonstrated the effectiveness of FVIII in the surgical treatment for type 3 VWD and further suggested that FVIII molecules are incorporated into platelet phospholipids to facilitate platelet activation as well as act directly to intrinsic pathways to normalize coagulation. Conclusions: Our observations suggested that FVIII plays an essential role in hemostasis in the absence of VWF and provided the rationale for the usage of rFVIII in the hemostatic management of type 3 VWD.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 628-628
Author(s):  
Grazia Loredana Mendolicchio ◽  
Reha Celikel ◽  
Kottayil I. Varughese ◽  
Brian Savage ◽  
Zaverio M. Ruggeri

Abstract Evaluation of the crystal structures of the amino terminal domain of platelet glycoprotein (GP) Ibα bound to the von Willebrand factor A1 domain (VWFA1) or to α-thrombin indicate the absence of significant steric hindrance in a putative triple complex of the two ligands interacting with the same receptor molecule. Superposition of the models reveals that intermolecular contacts may be established between VWFA1 and α-thrombin concurrently bound to GP Ibα, and suggests that these additional interactions could stabilize the intrinsically low affinity binding of the VWF A1 domain. To verify the predictions of the model, we used gel electrophoresis under native conditions and purified components in solution to demonstrate directly the formation of a triple complex. We then sought to evaluate whether α-thrombin could influence the functional effects of the VWF-GP Ibα interaction. For this purpose, we established a model of platelet agglutination/aggregation dependent on the interaction between recombinant dimeric VWFA1 domain, purified from the culture medium of stably transfected D. melanogaster cell lines, and GP Ibα. In this assay, platelet rich plasma prepared from individual donor blood collected with the thrombin inhibitor D-phenyl alanyl-L-prolyl-L-arginine chloromethyl ketone dihydrochloride (PPACK) as an anticoagulant (80 μM) was mixed with varying concentrations of dimeric VWFA1 (0.5-10 μg/ml) and exposed to variable shear rate levels in a cone-and-plate viscometer. Platelet aggregation was observed at shear rates between 6 and 108 dyn/cm2. The response in different normal controls was reproducible but variable in extent, and individuals could be assigned to one of two categories, low responder and high responder. An agglutination response was observed after platelets were treated with 10 μM prostaglandin E1 to block activation, and the distinction between low and high responders remained true under these conditions. For simplicity, agglutinated platelets were still defined as “aggregates”. With activation blocked platelets, aggregates were stable up to a shear rate of 30 dyn/cm2, but began to dissipate at higher levels. The addition of α-thrombin with the active site irreversibly blocked by PPACK at concentrations between 5 and 10 μg/ml substantially increased the extent of the platelet response. This was demonstrated by a faster rate of platelet agglutination/aggregation, a greater stability of aggregates at higher shear rates, and an overall increase in the size of aggregates formed. To demonstrate the latter, samples were exposed to shear stress under selected conditions and immediately fixed with 1% glutaraldehyde for quantitative image analysis. Maximum aggregate size was increased several fold in the presence of α-thrombin, and the difference was particularly evident in low responder individuals in whom dimeric VWFA1 alone caused the formation of small and unstable aggregates. PPACK-blocked thrombin by itself had no effect on platelet aggregate formation at any shear rate tested. Our findings delineate a mechanism through which α-thrombin may stabilize platelet-platelet contacts by mediating a tighter association between VWF A1 domain and GP Ibα receptor. Such a function, independent of proteolytic activity, may enhance platelet deposition at sites of vascular injury.


Blood ◽  
2014 ◽  
Vol 123 (1) ◽  
pp. 121-125 ◽  
Author(s):  
Julie Rayes ◽  
Lubka T. Roumenina ◽  
Jordan D. Dimitrov ◽  
Yohann Repessé ◽  
Mathieu Ing ◽  
...  

Key Points Complement factor H and von Willebrand factor colocalize in the Weibel-Palade bodies of endothelial cells and interact in normal plasma. Formation of the complex enhances FH cofactor activity and VWF-mediated platelet aggregation.


Blood ◽  
1990 ◽  
Vol 76 (2) ◽  
pp. 345-353 ◽  
Author(s):  
RR Hantgan ◽  
G Hindriks ◽  
RG Taylor ◽  
JJ Sixma ◽  
PG de Groot

Abstract We have investigated the molecular basis of thrombus formation by measuring the extent of platelet deposition from flowing whole blood onto fibrin-coated glass coverslips under well-defined shear conditions in a rectangular perfusion chamber. Platelets readily and specifically adhered to fibrin-coated coverslips in 5 minute perfusion experiments done at either low (300 s-1) or high (1,300 s-1) wall shear rates. Scanning electron microscopic examination of fibrin-coated coverslips after perfusions showed surface coverage by a monolayer of adherent, partly spread platelets. Platelet adhesion to fibrin was effectively inhibited by a monoclonal antibody (MoAb) specific for glycoprotein (GP) IIb:IIIa. The dose-response curve for inhibition of adhesion by anti-GPIIb:IIIa at both shear rates paralleled that for inhibition of platelet aggregation. Platelet aggregation and adhesion to fibrin were also blocked by low concentrations of prostacyclin. In contrast, anti- GPIb reduced adhesion by 40% at 300 s-1 and by 70% at 1,300 s-1. A similar pattern of shear rate-dependent, incomplete inhibition resulted with a MoAb specific for the GPIb-recognition region of von Willebrand factor (vWF). Platelets from an individual with severe von Willebrand's disease, whose plasma and platelets contained essentially no vWF, exhibited defective adhesion to fibrin, especially at the higher shear rate. Addition of purified vWF restored adhesion to normal values. These results are consistent with a two-site model for platelet adhesion to fibrin, in which the GPIIb:IIIa complex is the primary receptor, with GPIb:vWF providing a secondary adhesion pathway that is especially important at high wall shear rates.


2001 ◽  
Vol 85 (04) ◽  
pp. 679-685 ◽  
Author(s):  
Nancy Cauwenberghs ◽  
Agotha Schlammadinger ◽  
Stephan Vauterin ◽  
Susan Cooper ◽  
Gretel Descheemaeker ◽  
...  

SummaryIn this paper we describe two pathways leading to platelet activation by crosslinking glycoprotein (GP) Ib to the platelet Fc-receptor (FcγRII). First the monoclonal antibody (MoAb) 9C8, raised against human platelet GPIbα, dose-dependently induced platelet aggregation of citrate-anticoagulated platelet-rich plasma, an effect that can be inhibited by several activation inhibitors. The FcγRII-inhibitory MoAb IV.3 was able to prevent the aggregatory effects of MoAb 9C8, indicating that crosslinking of the antigen GPIbαto the FcγII-receptor is necessary for the activating effect. Secondly we observed a synergistic activating effect of two anti-von Willebrand factor (vWF) MoAbs 1C1E7 and B724, both known to enhance vWF binding to GPIbαin the presence of shear or ristocetin. When these antibodies are added together to PRP, platelet aggregation is induced without further need for an additional modulator. This effect can be blocked by either MoAb IV.3 or an inhibitory anti-GPIbαMoAb, indicating that again the platelet activation results from signaling through FcγRII crosslinked to vWF bound to GPIbα. In addition, both the anti-GPIbαMoAb 9C8, or the two anti-vWF MoAbs 1C1E7 and B724 induce genuine platelet activation, as evidenced by the secretion of ATP and protein tyrosine phosphorylation. These findings with both anti-GPIbαand anti-vWF MoAbs add further proof to recent reports demonstrating an interaction between the platelet receptors GPIbαand FcγRII, suggesting a role for the FcγII-receptor in GPIb-related signaling.


Sign in / Sign up

Export Citation Format

Share Document