Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood Advances, American Society of Hematology, Vol. 7, No. 7 ( 2023-04-11), p. 1258-1268
    Abstract: Platelet glycoprotein VI (GPVI) is attracting interest as a potential target for the development of new antiplatelet molecules with a low bleeding risk. GPVI binding to vascular collagen initiates thrombus formation and GPVI interactions with fibrin promote the growth and stability of the thrombus. In this study, we show that glenzocimab, a clinical stage humanized antibody fragment (Fab) with a high affinity for GPVI, blocks the binding of both ligands through a combination of steric hindrance and structural change. A cocrystal of glenzocimab with an extracellular domain of monomeric GPVI was obtained and its structure determined to a resolution of 1.9 Å. The data revealed that (1) glenzocimab binds to the D2 domain of GPVI, GPVI dimerization was not observed in the crystal structure because glenzocimab prevented D2 homotypic interactions and the formation of dimers that have a high affinity for collagen and fibrin; and (2) the light variable domain of the GPVI-bound Fab causes steric hindrance that is predicted to prevent the collagen-related peptide (CRP)/collagen fibers from extending out of their binding site and preclude GPVI clustering and downstream signaling. Glenzocimab did not bind to a truncated GPVI missing loop residues 129 to 136, thus validating the epitope identified in the crystal structure. Overall, these findings demonstrate that the binding of glenzocimab to the D2 domain of GPVI induces steric hindrance and structural modifications that drive the inhibition of GPVI interactions with its major ligands.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2023
    detail.hit.zdb_id: 2876449-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 114, No. 13 ( 2009-09-24), p. 2793-2801
    Abstract: We studied how integrin α2β1 and glycoprotein VI (GPVI) contribute to collagen-induced platelet activation under flow conditions by evaluating stable adhesion and intracellular Ca2+ concentration ([Ca2+]i) of FLUO 3-AM–labeled platelets perfused over acid-soluble type I or microfibrillar type VI collagen. Adhering platelets displayed 2 kinds of [Ca2+] i oscillations. Rapid α-like peaks were unaffected by the membrane-impermeable Ca2+ chelator ethyleneglycoltetraacetic acid but abolished by membrane-permeable BAPTA-AM. Longer-lasting γ-like peaks were always preceded by at least one α-like peak and abolished by intracellular or extracellular Ca2+ chelation. Inhibition of phosphatidylinositol 3-kinase or phospholipase C and modulation of cyclic nucleotides, but not blockage of adenosine diphosphate receptors, prevented both Ca2+ responses. Human or mouse platelets lacking GPVI function exhibited α-like but not γ-like Ca2+ peaks, whereas those lacking α2β1 showed markedly reduced to absent α-like and no γ-like Ca2+ peaks. Specific α2β1 ligation induced α-like but not γ-like peaks. Thus, α2β1 may generate Ca2+ signals that are reinforced by GPVI and required for subsequent longer-lasting Ca2+ oscillation mediated by GPVI through transmembrane ion flux. Our results delineate a GPVI-independent signaling role of α2β1 in response to collagen stimulation.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Society of Hematology ; 2010
    In:  Blood Vol. 116, No. 21 ( 2010-11-19), p. 3194-3194
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 3194-3194
    Abstract: Abstract 3194 In the blood, platelets are normally prevented from activation by endothelial inhibitors (i.e. prostacycline, ectonucleotidase). Dysfunctional endothelial cells loose their protective properties and favor platelet adhesion to matrix proteins, platelet aggregation and thrombus growth. Collagen fibers are highly thrombogenic and the platelet Glycoprotein (GP)VI predominantly mediates collagen-induced platelet responses. GPVI is a platelet specific receptor of the immunoglobulin (Ig) superfamily containing two extracellular Ig domains, a single transmembrane domain and a short cytoplasmic tail. GPVI signals through the immunoreceptor tyrosine-based activation motifs (ITAM) of the non-covalently associated immune receptor adaptor FcRg dimer. There is growing evidence that optimal binding of GPVI to collagen depends on the formation of GPVI dimers at the platelet surface: only dimeric GPVI binds to collagen and inhibits collagen-induced platelet aggregation and not monomeric GPVI. Moreover, crystallographic data showed dimerization of GPVI ectodomains. However, the valence of GPVI on resting and activated platelets is still debated. We have obtained an anti-human GPVI monoclonal antibody (9E18), that binds to dimeric GPVI with a 200 fold higher affinity than to monomeric GPVI. In flow cytometry on whole blood, while the 3J24 antibody labels 〉 95% platelets, 9E18 hardly binds to resting platelets with less than 3% positive platelets. The level of 9E18-positive platelets moderately increased (10-15%) after platelet isolation suggesting it could reflect platelet activation. Binding of 9E18 was indeed significantly increased on ADP- or TRAP-activated washed platelets (25±1.9 % and 36±7% positive platelets respectively). Additionally, increased binding of 9E18 was triggered by the GPVI agonists, collagen, convulxin or the activating 9O12 IgG. At sites of vascular lesion, platelet adhesion is initiated by the shear-dependent interaction of GPIb with vWF, assumed to favor GPVI-collagen interaction. When a platelet rich plasma was submitted to a shear of 4000 s-1 for 5 min, 9E18-positive platelets increased from 3.6±1.6% to 7±2% in the whole platelet population and to 26±7.7% on small aggregates (p 〈 0.05).When a2b1 and aIIbb3 were blocked, the relation between the 9E18 binding to stimulated platelets and platelet binding to collagen was linear (r2 = 0.847, p=0.0012, n=8). Interestingly, the cAMP elevating agent PGE1 further lowered the level of 9E18-binding to resting platelets and dropped it to basal values on ADP- or TRAP-treated platelets. Apyrase reduced by 50% TRAP-induced binding of 9E18 whereas indomethacin had no effect. PMA triggered binding of 9E18 on platelets (p 〈 0.001) while the Tyr-phosphatase inhibitor PAO, strongly inhibited PMA-induced 9E18 binding to platelets (p 〈 0.0019) and GPVI-dependent platelet adhesion to collagen. Altogether, these data indicate that 9E18 permit to quantify GPVI dimers on platelets. They show that (i) GPVI is mainly monomeric on resting platelets, (ii) dimerisation is an active process triggered by shear, soluble agonists and matrix proteins, (iii) the level of GPVI dimers is related to the capacity of platelets to adhere to collagen, (iv) GPVI dimerisation is completely prevented in the presence of agents increasing cAMP or by PAO. These data suggested that the formation of GPVI dimer is strictly controlled on resting platelets and that GPVI dimers could thus represent a new marker of platelet activation and susceptibility to collagen. Indeed, in a population of hospitalized patient, a positive correlation was observed between 9E18 binding and P-selectin exposure on platelets. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Society of Hematology ; 2014
    In:  Blood Vol. 124, No. 21 ( 2014-12-06), p. 4152-4152
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 4152-4152
    Abstract: Background: Fibrin, the end product of the coagulation cascade, consolidates the platelet plug at site of thrombosis: polymerized fibrin supports platelet adhesion under low and high shear rate conditions (Hantgan RR et al., Thromb Haemost 1992) and triggers platelet procoagulant activity (Beguin S et al., Blood 1999). These responses are largely independent of the integrin αIIβ3 and are carried by a yet ill-defined receptor. Platelet glycoprotein VI (GPVI) has a well-established key role in the initiation of thrombosis since it supports collagen-mediated platelet activation but it has recently been recognized to interact with other macromolecules such as fibronectin, vitronectin and laminins. We hypothesized GPVI could be the “missing” platelet receptor of fibrin. Aim of the study: to challenge the hypothesis that glycoprotein VI (GPVI) could be a functional fibrin receptor Methods: Thrombin generation was measured using calibrated automated thrombogram (CAT) in PRP from healthy volunteers, four GPVI-deficient patients and one patient with a fibrinogen deficiency. CAT was also performed on washed platelets mixed with prothrombin complex (FII, FVII, FIX, FX), antithrombin and fibrinogen. GPVI was blocked using the Fab of the monoclonal antibody 9O12. Fibrin polymerization was blocked using the GPRP peptide. GPVI binding to fibrin was measured in vitro using recombinant soluble GPVI (GPVI-Fc). Flow based adhesion assays were performed in capillary chambers coated with polymerized fibrin at variable shear rates and platelet morphological changes analyzed by scanning electron microscopy. The formation of fibrin-platelet thrombi was visualized by perfusing recalcified blood containing A647 fibrinogen in flow chambers (Vena8 Fluoro+ Cellix) coated with collagen and tissue factor. In a second step, the perfusion of hirudinated blood in which platelets were stained by A488-RAM1 allowed to visualize platelet recruitment by fibrin rich clots. Results: Thrombin generation triggered by tissue factor was impaired in the PRP of patients with a GPVI deficiency or in the presence of the Fab 9O12 as indicated by a respective decrease in the peak height of 45 and 25% as compared to controls. This effect was observed regardless the trigger of thrombin generation and required platelet activation. Measuring thrombin generation in a purified system showed that fibrinogen dose-dependently increased the thrombin peak by up to 150% at 3 mg/mL but the Fab 9O12 blunted this effect. Moreover, the Fab 9O12 had no effect on thrombin generation in the PRP of a fibrinogen-deficient patient confirming a GPVI/fibrin(ogen)interplay. Blocking fibrin polymerization by GPRP reduced the thrombin peak in normal PRP, in fibrinogen-supplemented PRP of the fibrinogen-deficient patient and in purified conditions. In contrast GPRP had no effect on the thrombin peak in normal PRP containing the Fab 9O12 and in the PRP of GPVI-deficient patients. The proof that GPVI specifically interacts with fibrin was obtained in a binding assay showing a dose-dependent binding of GPVI-Fc to fibrin polymers that was reversed by the Fab 9O12. Platelets adhered to polymerized fibrin resulting in platelet shape change and exposure of phosphatidylserine. Platelet adhesion on a fibrin network was observed at low (300 s-1) and high (1500 s-1) shear rates with the formation of small contractile thrombi. Adhesion was decreased by 62% for 9O12-treated platelets and by 43% with the blood of GPVI-deficient mice as compared to controls. Importantly, lack of GPVI or its blockade decreased stationary adhesion indicating that GPVI is required to stabilize the interactions between platelets and fibrin. Finally when hirudinated blood was perfused at a shear rate of 1500 s-1 onto preformed fibrin-rich clots, the Fab 9O12 decreased the recruitment of platelets by up to 93%. Conclusions: Here we show for the first time that GPVI acts as a receptor for polymerized fibrin with two major functions: GPVI interaction with polymerized fibrin triggers (i) a new loop amplifying thrombin generation and (ii) platelet recruitment at the clot surface. These, so far, unrecognized properties of GPVI confer it a key role in the maturation of the thrombus by facilitating its growth and stabilization in addition to its well-known effect in the initiation of thrombus formation. Disclosures Jandrot-Perrus: Acticor Biotech: Other. Gachet:Acticor Biotech: Other.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Society of Hematology ; 2011
    In:  Blood Vol. 118, No. 21 ( 2011-11-18), p. 618-618
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 618-618
    Abstract: Abstract 618 Disorders of hemostasis are frequent in patients suffering from classic myeloproliferative neoplasm (MPN) i.e. essential thrombocytemia (ET) or polycythemia vera (PV). They include high incidence of thrombosis, which represents a leading cause of morbidity and mortality, and a lower incidence of hemorrhagic events. The pathogenesis of hemostatic disorders is complex. The JAK2V617F mutation is found in 95% of PV and in 50% of ET. Since receptors coupled to JAK2V617F are hyper-responsive to growth factors, it is proposed that JAK2V617F primes platelets and, as a consequence, directly contributes to the thrombotic risk. However, the relation between the mutation and thrombosis has yet not clearly been established due to variable JAK2V617F allele burden and antithrombotic prophylaxis. Novel MPN animal models expressing the mutation have been developed with reported hemostasis disorders from unknown origin. Therefore, we took the opportunity to use these JAK2V617F knock-in (KI) mice to investigate the effects of JAK2V617F on platelet functions. We used irradiated WT recipient mice grafted with bone marrow (BM) cells from JAK2+/V617F KI donor mice. JAK2+/V617F KI mice were conditional floxed JAK2+/fl mice crossed with transgenic mice expressing Cre under the control of the vav promoter (vavCre/JAK2+/V617F) or Cre-tamoxifen-inducible transgenic mice expressing CreERt under the HSC-SCL promoter (sclCreERt/JAK2+/V617F). All recipient mice developed 4 weeks after transplantation (vavCre/JAK2+/V617F) and tamoxifen-induction (sclCreERt/JAK2+/V617F) a MPN mimicking PV and characterized by elevated hematocrit, platelet and leucocyte levels. Mice were studied between 4 and 9 weeks after the graft or 7, 14, 32 and 60 days after tamoxifen-induction (sclCreERt/JAK2+/V617F). Platelet functions were analyzed in vitro by flow cytometry and light transmission aggregometry, ex vivo by measuring thrombus formation in whole blood and flow conditions on collagen and vWF, and in vivo in a FeCl3-induced model of thrombosis. The analysis of platelet glycoprotein expression was focused on two ITAM receptors, GPVI and CLEC2. The principal observation is a deficiency in GPVI up to 50% in vavCre/JAK2+/V617F mice (p 〈 0.01) while expression of other platelet membrane glycoproteins (GPIb, GPV, GPIIbIIIa and CLEC2) was normal. GPVI deficiency was confirmed by immunoblot while the expression of the FcRg chain was normal. At arterial shear rates, platelet adhesion and aggregation on immobilized collagen and vWF were profoundly impaired with a platelet surface coverage decreased by up to 80% (p 〈 0.01). In vivo, an enlargement of mesenteric vessels was noticed in vavCre/JAK2+/V617F mice. When vessels of the same caliber as WT mice were submitted to FeCl3-induced thrombosis, platelet aggregates formed rapidly (5-8 min) but surprisingly occlusion was not often reached and the aggregates were highly unstable. In sclCreERt/JAK2+/V617F mice, GPVI deficiency was not observed at day 7 and 14 but appeared at day 60 after JAK2V617F-induction by tamoxifen. In vivo, time to vessel occlusion and thrombus stability were initially (day 7 and 14) similar in KI and WT mice. However, the rate to which platelet aggregates formed and their stability became impaired at 32 days after JAK2V617F-induction. In conclusion, these data demonstrate that JAK2V617F induces complex modifications of platelet functions including platelet GPVI deficiency, in vitro loss of collagen-induced aggregation and facilitation of in vivo thrombosis initiation associated with instability of the clot. Therefore, this study provides new insight into the pathophysiology of hemostatic disorders in MPN, thrombus instability being able to drive microvascular thrombosis disorders and pulmonary embolism. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 3638-3638
    Abstract: We have investigated signaling mediated by GPIb-IX-V, GPVI and α2β1 during platelet adhesion to collagen type I either in the presence or absence of von Willebrand factor (VWF) A1A2A3 domain under flow conditions (wall shear rate 600 or 3000 s−1). We used platelets labeled with FLUO3-AM and real-time videomicroscopy with high-speed image acquisition to analyze concurrently adhesion, translocation and calcium transients in single platelets interacting with the surface. All experiments were performed in the presence of integrillin (100 μg/ml) to block the possible involvement of αIIbβ3. Platelet adhesion and activation at 600 s−1 were similar in the absence or presence of VWF A1A2A3 domain. Calcium signaling consisted of both short lasting peaks (release from intracellular stores) and long sustained waves (transmembrane flux). In our experimental condition blockade of α2β1 or GPVI with monoclonal antibodies reduced platelet adhesion by 50% and the proportion of adherent platelets that became activated by 20–30%. Concomitant inhibition of both receptors decreased platelet adhesion by & gt;90%. Thus, both α2β1 and GPVI participate in the initial stage of platelet adhesion to collagen type I. At the higher shear rate of 3000 s−1, platelet adhesion and activation was 40–50% less than at 600 s−1 in the absence of VWF A1A2A3 domain; addition of the latter enhanced adhesion and activation by 12 to 14-fold, and & gt;80% of the platelets exhibited stable adhesion during the 30 s observation period. Intracellular Ca++ peaks in adherent platelets reached 2–3 μM. Blockade of GP Ib-IX-V inhibited both platelet adhesion and activation by at least 90%. Blockade of α2β1 markedly reduced the number of adhering platelets, which were mostly ( & gt;90%) translocating on the surface and exhibited only short lasting Ca++ transients. Blockade of GPVI greatly decreased the number of adherent platelets, most of which, however, were firmly attached to the surface ( & lt;15% exhibiting translocation). Calcium transients in the activated platelet consisted of short lasting peaks and rare long lasting waves reaching concentrations & lt;1.5–2 μM. These results suggest that, on a surface of collagen type I, α2β1 is required for normal platelet arrest following tethering through the VWF A1 domain-GPIb-IX-V interaction at higher shear rates, and Ca++ signaling results from the concerted action of the two receptors reinforced by GPVI. The adhesion potential of platelets exposed to collagen, therefore, appears to depend on signaling triggered by different ligand-receptor interactions varying as a function of flow conditions.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood Advances, American Society of Hematology, Vol. 2, No. 18 ( 2018-09-25), p. 2389-2399
    Abstract: Coagulation and fibrinolytic system deregulation has been implicated in the development of idiopathic pulmonary fibrosis, a devastating form of interstitial lung disease. We used intratracheal instillation of bleomycin to induce pulmonary fibrosis in mice and analyzed the role of serine protease inhibitor E2 (serpinE2)/protease nexin-1 (PN-1), a tissue serpin that exhibits anticoagulant and antifibrinolytic properties. PN-1 deficiency was associated, after bleomycin challenge, with a significant increase in mortality, as well as a marked increase in active thrombin in bronchoalveolar lavage fluids, an overexpression of extracellular matrix proteins, and an accumulation of inflammatory cells in the lungs. Bone marrow transplantation experiments showed that protective PN-1 was derived from hematopoietic cell compartment. A pharmacological strategy using the direct thrombin inhibitor argatroban reversed the deleterious effects of PN-1 deficiency. Concomitant deficiency of the thrombin receptor protease-activated receptor 4 (PAR4) abolished the deleterious effects of PN-1 deficiency in hematopoietic cells. These data demonstrate that prevention of thrombin signaling by PN-1 constitutes an important endogenous mechanism of protection against lung fibrosis and associated mortality. Our findings suggest that appropriate doses of thrombin inhibitors or PAR4 antagonists may provide benefit against progressive lung fibrosis with evidence of deregulated thrombin activity.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 2876449-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 96, No. 5 ( 2000-09-01), p. 1798-1807
    Abstract: Injuries to the vessel wall and subsequent exposure of collagen from the subendothelial matrix result in thrombus formation. In physiological conditions, the platelet plug limits blood loss. However, in pathologic conditions, such as rupture of atherosclerotic plaques, platelet–collagen interactions are associated with cardiovascular and cerebral vascular diseases. Platelet glycoprotein VI (GPVI) plays a crucial role in collagen-induced activation and aggregation of platelets, and people who are deficient in GPVI suffer from bleeding disorders. Based on the fact that GPVI is coupled to the Fc receptor (FcR)-γ chain and thus should share homology with the FcR chains, the genes encoding human and mouse GPVI were identified. They belong to the immunoglobulin (Ig) superfamily and share 64% homology at the protein level. Functional evidence demonstrating the identity of the recombinant protein with GPVI was shown by binding to its natural ligand collagen; binding to convulxin (Cvx), a GPVI-specific ligand from snake venom; binding of anti-GPVI IgG isolated from a patient; and association to the FcR-γ chain. The study also demonstrated that the soluble protein blocks Cvx and collagen-induced platelet aggregation and that GPVI expression is restricted to megakaryocytes and platelets. Finally, human GPVI was mapped to chromosome 19, long arm, region 1, band 3 (19q13), in the same region as multiple members of the Ig superfamily. This work offers the opportunity to explore the involvement of GPVI in thrombotic disease, to develop alternative antithrombotic compounds, and to characterize the mechanism involved in GPVI genetic deficiencies.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2000
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3200-3200
    Abstract: Background: Myeloproliferative neoplasms (MPN) are associated with an increased risk of arterial and venous thrombosis with an annual incidence varying from 1.1% to 6.6% (Patrono, Blood 2013). Comparable to that recommended in non-MPN high-risk patients, low-dose aspirin (81-100 mg/day) is recommended in primary and secondary prophylaxis of thrombosis in polycythemia vera and essential thrombocytemia (Tefferi, Am J Hematol 2012). However, the daily dose of aspirin to optimize efficacy and safety for MPN patients has never been challenged. Aim of the study: Determine the optimal dose of aspirin to achieve biological efficacy in Philadelphia-negative MPN patients. Methods: Patients with Philadelphia-negative MPN who were treated with low-dose aspirin 75 or 100 mg/day were enrolled in this observational study. Patients without any cytostatic drug and patients treated with either pegylated interferon alpha or hydroxyurea were eligible. Major exclusion criteria included inability to adhere to aspirin therapy and chronic oral anticoagulation. Biological efficacy to aspirin was evaluated by platelet aggregation induced by arachidonic acid 1.33 mM on platelet rich plasma and tested at trough level and 24 hours after last aspirin intake. Resistance to aspirin was defined as a maximal platelet aggregation over 20%. According to the results of platelet aggregation, aspirin dose and dosing regimen were modified as follows: from 100 mg/day to 160 mg/day or 75 mg x2/day. Patients enrolled in this have a median follow up of at least 6 months after the analysis. Results: Between January 2012 and February 2014, 77 patients with Philadelphia-negative MPN were included. 53 were treated with aspirin 75 mg/day (69%) and 24 with aspirin 100 mg/day (31%). Out of the 53 patients treated with aspirin 75 mg/day, 12 patients (23%) were resistant to aspirin. Resistance to aspirin was not correlated to sex, age, presentation, JAK2 status, treatment, history of thrombotic or bleeding and hematologic values (see following table). Table 1:The MPN Grade 1 Fibrosis PhenotypePMF (N:33)PV/ET (N:58)Total (N:91)Median age63 yrs55 yrs58 yrsSex1:31:11:2JAK V617F +16/33: (49%)40/58: (69%)56/91: (62%)Median Hgb(g/dL)11.9 (range 7.9-16.4)12.5 (range 8.0-19.8)12.2 (range 7.9-19.8)Median WBC(X10 (9))26.9 (range 1.3-188)8.9 (range 3.5-51.3)10.8 (range 1.3-188 )Median Platelet(X10 (9))179 (range 18.0-1194)505 (range 67-2286)370 (range 18-2286)Leukoerythroblastic Blood Smear15/33 (45%)PV: 17 ET: 5 Total: 22/58 (38%)37/91 (41%)Splenomegaly(cm below costal margin)18/33: (55%) Median: 1025/58: (43%) Median: 643/91(48%) Median: 4Transfusion dependence6/33: (18%)2/58: (4%)8/91: (9%)Presence Of ³ 1 symptom 17/33: (52%)33/58: (57%)50/91: (55%)DIPSS risk intermediate 2 or higher13/33: (39%)17/58: (29%)30/91: (33%)2 or more prior therapies9/33: (27%)36/58: (62%)45/91: (49%)Vital Status (Alive)25/33: (76%)53/58: (91%)78/90: (86%)Median follow up time (yrs)1.9 (range 0.1-9.8)5.7 (range 0-34.5)3.1 (range 0-34.5) An increased dose of 100 mg/day for at least 7 days overcame this biological resistance in 8 out of 8 re-tested patients. The 2 remaining 75mg resistant patients received an increased dose of aspirin but were not retested. Out of the 24 patients under 100 mg/day, only two (8%) were resistant to aspirin. In these patients, increasing the dose to 160 mg/day did not modify their biological response. However a 75 mgx2 /day was effective to overcome biological resistance. No thrombotic or bleeding event was observed during the 6-month follow-up regardless of the aspirin dose. Conclusions: This is the first study to measure in standardized conditions the biological resistance of aspirin in 77 well-characterized Philadelphia-negative MPN patients. Although this does not reach statistical significance a higher proportion of patients treated with aspirin 75mg/day was resistant to anti-platelet therapy compared to patients treated with aspirin 100 mg/day. Increasing the dose of aspirin from 75 to 100 mg once daily overcomes this biological resistance without increasing bleeding side effects and seems the best compromise. Interestingly in rare cases of extreme resistance to 100mg doubling the dose twice daily (75mgX2) was better than an increase of 160mg in one take. This pilot study on a small number of patients with a limited 6-month follow up compared to the low annual incidence of thromboses in these MPNs will be followed by a prospective study on a larger number of patients with an extended follow-up period to determine if biological resistance to aspirin is correlated to the occurrence of thrombotic events. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Society of Hematology ; 2022
    In:  Blood Vol. 139, No. 20 ( 2022-05-19), p. 3005-3007
    In: Blood, American Society of Hematology, Vol. 139, No. 20 ( 2022-05-19), p. 3005-3007
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages