Potency Adjusted Generic Versions of Argatroban Can Be Differentiated from Branded Argatroban in Thrombin Generation and Platelet Activation Assays.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4007-4007
Author(s):  
Cafer Adiguzel ◽  
Omer Iqbal ◽  
Michael Sammikannu ◽  
Josephine Cunanan ◽  
Walter Jeske ◽  
...  

Abstract Argatroban represents a widely used direct parenteral thrombin inhibitor for the anticoagulation management of patients with heparin-induced thrombocytopenia. Several generic versions of argatroban namely Slovastan, Gartban and Argaran have also become available in Japan. Although the antithrombin potency of the generic products of argatroban is adjusted to be comparable to the branded product, apparent differences in the pharmacodynamic effects have been noted in thrombin generation and platelet activation assays. To further investigate the bioequivalence of the three generic products with the branded argatroban, these agents were compared in whole blood (WB), platelet rich plasma (PRP) platelet poor plasma (PPP) and isolated biochemical systems. In the WB assays, the activated clotting time (ACT) studies were carried out mimicking the anticoagulant dosing (0–5 ug/ml). In the citrated WB, PRP and PPP various clotting tests such as the prothrombin time/INR (PT/INR), activated partial thromboplastin time (APTT), Heptest, prothrombinase activated clotting time (PICT) and thrombin time were carried out. To test the effect of these agents on tissue factor mediated activation of blood cells, flow cytometric studies were carried out. In addition, thrombin generation markers such as the fibrinopeptide A, thrombin/antithrombin complex and prothrombin fragment 1.2 were also measured. The effect of different forms of argatroban were also investigated on Xa and thrombin generation inhibition. While there was no difference in the anticoagulant effects of the branded and generic products in the clotting assays such as the PT, APTT, Heptest, PICT and thrombin time, matrix based differences were apparent. In the ACT assay, the anticoagulant effect of the branded and generic product were approximately the same, however, upon supplementation of the tissue factor the relative anticoagulant effects of these agents differed. All of the agents also produced a concentration dependent inhibition of the generation of microparticles in the WB studies where each of these agents were differentiated. Argaran produce weaker responses than the other agents. All of the agents also blocked p-selectin expression induced by tissue factor with an IC50 ranging from 1.8–2.3 ug/ml. There were obvious differences among the generic and branded products. In the thrombin and Xa generation assays differences were also noted between the generic and branded product. The relative ability of the generic and the branded argatroban in inhibiting the activation of thrombin activatable fibrinolysis inhibitor (TAFI) showed noticeable differences. These studies clearly indicated that while in the antithrombin titration and global anticoagulant assays the generic brands of argatroban exhibit comparable effects, in cellular systems and other assays differences between the generic product and branded versions can be noted. These obvious differences may be related to the solution matrix and the relative proportion of different forms of argatroban. These observations warrant additional pharmacoequivalent studies on the generic product to assure clinical equivalence of these products.

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3207-3207
Author(s):  
Patrick Van Dreden ◽  
Joseph Gligorov ◽  
Evangelos Terpos ◽  
Mathieu Jamelot ◽  
Michele Sabbah ◽  
...  

Abstract Background: COVID-19 has been associated with hypercoagulability, endothelial cell injury and frequent thrombotic complications resulting both from direct effects of the virus on the endothelium and from the 'cytokine storm' resulting from the host's immune response. Since the COVID-19 vaccines have been shown to effectively prevent symptomatic infection including hospital admissions and severe disease, the risk of COVID-19-related thrombosis should be expected to (almost) disappear in vaccinated individuals. However, some rare cases of venous thrombosis have been reported in individuals vaccinated with mRNA vaccines. Thus, there is a sharp contrast between the clinical or experimental data reported in the literature on COVID-19 and on the rare thrombotic events observed after the vaccination with these vaccines. This phenomenon raised some scepticism of even some fear about the safety of these vaccines which could compromise the adhesion of the citizens in the vaccination program. Aims: We conducted a prospective observational study, to explore the impact of vaccination with the BNT162b2 (Pfizer/BioNTech) on blood hypercoagulability and endothelial cell activation and to investigate if this is modified by the presence of active cancer. Methods: In total 229 subjects were prospectively included in the study from April to June 2021. Subjects were stratified in three predefined groups: 127 vaccinated patients with active cancer (VOnco group), 72 vaccinated health care workers (VHcw group) and 30 non vaccinated health individuals (Control group). Blood samples were obtained 2 days after the administration of the first dose of BNT162b2 vaccine and collected in Vacutainer® tubes (0.109 mol/L trisodium citrate). Platelet poor plasma (PPP) was prepared by double centrifugation at 2000 g for 20 minutes at room temperature and plasma aliquots were stored at -80°C until assayed. Samples of PPP were assessed for thrombin generation (TG) with PPP-Reagent® (Thrombogram-Thrombinoscope assay with PPP-Reagent®TF 5pM), E-selectin, D-dimers, (D-Di), Tissue Factor (TFa), procoagulant phospholipid-dependent clotting time (Procag-PPL) and von Willebrand factor (vWF), thrombomodulin (TM), tissue factor pathway inhibitor (TFPI), and platelet factor 4 (PF4). All assays were from Diagnostica Stago (France). The upper and lower normal limits (UNL and LNL) for each biomarker were calculated by the mean±2SD for the control group. Results: All vaccinated subjects showed significantly increased levels of PF4 (71% >UNL, p<0.001), D-Dimers (74% >UNL, p<0.01), vWF (60% >UNL, p<0.01), FVIII (62% >UNL, p<0.01) and shorter Procoag-PPL clotting time (96% <LNL, p<0.001), as compared to controls. Thrombin generation showed significantly higher Peak (60% >UNL, p<0.01), ETP (38% >UNL, p<0.01) and MRI (66% >UNL, p<0.01) but no differences in lag-time in vaccinated subjects as compared to the control group. Vaccinated subjects did not show any increase at the levels of TFa, TFPI, TM and E-selectin in comparison with the control group. The studied biomarkers were not significantly different between the VOnco and VHcw groups. Conclusion: The ROADMAP-COVID-19-Vaccine study shows that administration of the first dose of the BNT162b2 vaccine induced significant platelet activation documented by shorter Procoag-PPL associated with increased levels of PF4. Plasma hypercoagulability was less frequent in vaccinated individuals whereas there was no evidence of significant endothelial cells activation after vaccination. Interestingly, the presence of active cancer was not associated with an enhancement of platelet activation, hypercoagulability, or endothelial cell activation after the vaccination. Probably, the generated antibodies against the spike protein or lead to platelet activation in a FcyRIIa dependent manner that results in PF4 release. The implication of the mild inflammatory reaction triggered by the vaccination could be another possible pathway leading to platelet activation. Nevertheless, vaccination does not provoke endothelial activation even in patients with cancer. The findings of the ROADMAP-COVID-19-Vaccine study support the concept administration of mRNA based vaccines does not directly cause a systematic hypercoagulability. Disclosures Gligorov: Roche-Genentech: Research Funding; Novartis: Research Funding; Onxeo: Research Funding; Daichi: Research Funding; MSD: Research Funding; Eisai: Research Funding; Genomic Heatlh: Research Funding; Ipsen: Research Funding; Macrogenics: Research Funding; Pfizer: Research Funding. Terpos: Novartis: Honoraria; Janssen: Consultancy, Honoraria, Research Funding; Genesis: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria, Research Funding; BMS: Honoraria; Amgen: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria, Research Funding; Sanofi: Consultancy, Honoraria, Research Funding; GSK: Honoraria, Research Funding. Dimopoulos: Amgen: Honoraria; BMS: Honoraria; Janssen: Honoraria; Beigene: Honoraria; Takeda: Honoraria.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 912-912 ◽  
Author(s):  
Bernd Jilma ◽  
Judith M. Leitner ◽  
Francesco Cardona ◽  
Florian B. Mayr ◽  
Christa Firbas ◽  
...  

Abstract Background: BIBT 986 is a novel potent anticoagulant that dually inhibits Factors Xa and IIa. We hypothesized that BIBT 986 would dose-dependently decrease endotoxin-induced, tissue factor triggered coagulation activation. Hence it was the aim of the study to compare with placebo the anticoagulant activity of three dosages of BIBT 986 on parameters of coagulation, platelet activation and inflammation and to examine the safety of BIBT 986 in this setting. Methods: This study was a prospective, randomized, double-blind, placebo-controlled, parallel-group dose escalation trial in 48 healthy male volunteers. Participants were randomised to receive bolus primed continuous infusions of one of the three doses of BIBT 986 or placebo. All of them received a bolus infusion of 2ng/kg body weight lipopolysaccharide (LPS). Results: BIBT dose-dependently increased anti-Xa activity, activated partial thromboplastin time (APTT), ecarin clotting time (ECT), thrombin time (TT) and the international normalisation ratio (INR). Importantly, BIBT 986 dose-dependently blocked the LPS-induced coagulation as assessed by the in vivo markers of thrombin generation and action: BIBT 986 doses that prolonged APTT by 25% were already effective. The BIBT dose that prolonged APTT by 100%, completely suppressed the increase in prothrombin fragment (F1+2), thrombin-antithrombin complexes (TAT) and D-dimer. BIBT 986 had no influence on activation markers of inflammation, fibrinolysis, endothelial or platelet activation. Conclusion: Infusion of BIBT 986 was safe and well tolerated. BIBT 986 specifically and dose-dependently blocked LPS-induced, tissue factor trigger coagulation. When compared to different anticoagulants tested previously in this standardized model, BIBT 986 was more effective in suppressing thrombin generation (F1+2 levels) than standard doses of danaparoid, dalteparin or lepirudin. BIBT 986 represents the first drug of a new class of dual FXa and FIIa inhibitors, and displays high potency.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 5129-5129
Author(s):  
Jawed Fareed ◽  
Debra Hoppensteadt ◽  
Omer Iqbal ◽  
Jeanine M. Walenga ◽  
Bruce E Lewis

Abstract Abstract 5129 Several generic versions of argatroban) (Mitsubishi; Tokyo, Japan) have been introduced in Japan (Argaron, Gartban, Slovastan). In addition, other generic versions of argatroban are being considered by the European and North American regulatory bodies. While the generic versions of argatroban exhibit similar antithrombin potency (Ki values), because of the differential compositional variations their anticoagulant effects in whole blood systems may differ due to their cellular and plasmatic protein interactions. Branded and generic versions of argatroban may exhibit differential anticoagulant actions in the whole blood and plasma based assays due to their differential interactions with blood cells, platelets and plasma proteins. Three generic versions of argatroban that are commercially available in Japan namely Argaron, Gartban and Slovastan and a powdered version of generic argatroban (Lundbeck) were compared with the branded argatroban. Native whole blood thrombelastographic (TEG) analysis was carried out at 0.1 ug/mL, the Activated Clotting Time (ACT) assay was carried out in a concentration range of 0–10 ug/mL, and such coagulation tests as the PT/INR, aPTT, Heptest, and calcium thrombin time were performed. Plasma retrieved from the supplemented whole blood was also assayed. Ratios of the clotting time test values from whole blood and plasma were calculated. Retrieved plasma samples were also assayed in the thrombin generation assays (TGA). All of the different versions of argatroban produced a concentration dependent anticoagulant effect in the native whole blood TEG and ACT. In the TEG, while argatroban and Slovastan showed a similar effect, Gartban, Argaron and a powdered generic showed weaker effects. Argatroban was also different in the ACT assay. At a concentration of 5 ug/ml the ACTs were, Arg 340+15.2 secs, S 297+10.5 secs, G 292.0+19.1 secs and A 285.2+21.7 secs. In the citrated whole blood systems, all agents produced a concentration dependent anticoagulant effect; however, the generic versions produced a stronger anticoagulant effect in comparison to branded argatroban (p<0.001). In the PT assay at 5 ug/mL, argatroban showed 32 ± 3 sec vs 40–50 sec for the generic products. Similarly in the aPTT, Heptest and thrombin time tests argatroban was weaker than the generic products. Differences among generic versions were also evident. Similar results were obtained in the retrieved plasma, however the ratio of whole blood over plasma varied from product to product. The IC50 of the generic and branded argatrobans in the TGA were also different. These results show that while in the thrombin inhibition assays generic and branded argatroban may show similar effects, these agents exhibit assay dependent differences in the whole blood and plasma based assays. Such differences may be more evident in the in vivo studirs where endothelial cells and other interactions may contribute to product individuality. Therefore, based on the in vitro antiprotease assays, generic argatrobans may not be considered equivalent and require a multi-parametric study. Currently available generic argatrobans may not be equivalent in the in vivo anticoagulant effects. Therefore, clinical validation of the clinical equivalence for these drugs is warranted. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 26 ◽  
pp. 107602961989512
Author(s):  
Fakiha Siddiqui ◽  
Alfonso Tafur ◽  
Emily Bontekoe ◽  
Omer Iqbal ◽  
Walter Jeske ◽  
...  

Andexanet alfa is a recombinant factor Xa decoy protein, designed to reverse bleeding associated with oral anti-Xa agents. Andexanet alfa is also reported to neutralize the effects of heparin-related drugs. This study focused on the neutralization profiles of unfractionated heparin (UFH), enoxaparin, and, a chemically synthetic pentasaccharide, fondaparinux by andexanet alfa. Whole blood clotting studies were carried out using thromboelastography (TEG) and activated clotting time (ACT). The anticoagulant profile of UFH, enoxaparin, and fondaparinux was studied using the activated partial thromboplastin time (aPTT), thrombin time (TT), and amidolytic anti-Xa, and anti-IIa methods. Thrombin generation inhibition was studied using the calibrated automated thrombogram system. Reversal of each of these agents was studied by supplementing andexanet alfa at 100 µg/mL. In the TEG, andexanet alfa produced almost a complete reversal of the anticoagulant effects of UFH and enoxaparin; however, it augmented the effects of fondaparinux. In the ACT, aPTT, and TT, UFH produced strong anticoagulant effects that were almost completely neutralized by andexanet alfa. Enoxaparin produced milder anticoagulant responses that were partially neutralized, whereas fondaparinux did not produce any sizeable effects. In the anti-Xa and anti-IIa assays, UFH exhibited partial neutralization whereas enoxaparin and fondaparinux did not show any neutralization. All agents produced varying degrees of the inhibition of thrombin generation, which were differentially neutralized by andexanet alfa. These results indicate that andexanet alfa is capable of differentially neutralizing anticoagulant and antiprotease effects of UFH and enoxaparin in an assay-dependent manner. However, andexanet alfa is incapable of neutralizing the anti-Xa effects of fondaparinux.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 908-908 ◽  
Author(s):  
Walter P. Jeske ◽  
Jeanine M. Walenga ◽  
Paul D. Ackerman ◽  
Debra A. Hoppensteadt ◽  
Curtis Vandenberg ◽  
...  

Abstract Currently several generic versions of a branded low molecular weight heparin (LMWH; enoxaparin, Sanofi-Aventis, Paris, France) have become available for clinical use in several countries. Such products include Cutenox (Gland Pharma, Hyderabad, India), Dripanina (Ariston, São Paulo, Brazil) and Clenox (Pharmayect, Barranquilla, Columbia). Although these products are not currently approved for use in the U.S., several other manufacturers have sought FDA approval for their products. Due to a lack of specifications and guidelines, this approval is still pending. In order to compare the relative potency of different anti-factor Xa U/ml adjusted generic preparations, studies were designed to compare each of the individual generic LMWHs with the branded product. Additionally, multiple batches of some of the generic products were also profiled. All of the agents were tested in human whole blood and citrated plasma over a concentration range of 0.15 to 10 U/ml. Whole blood activated clotting time (ACT) and thrombelastography (TEG) measurements along with fibrinopeptide A (FPA) generation were compared. The plasmatic tests included anti-Xa and anti-IIa activity by amidolytic assay and aPTT, Heptest and PiCT clotting time assays. In addition, protamine sulfate neutralization profiles for these agents were investigated at fixed protamine concentrations of 12.5 and 25 μg/ml. In the whole blood assays, at concentrations &lt; 2.5 U/ml, no significant differences were observed between the branded and potency adjusted generic LMWHs. However, in the plasma-based systems, assay-dependent variations were observed which were more obvious at concentrations &gt; 1.25 U/ml (aPTT, anti-IIa, anti-Xa; p&lt;0.05). Similarly, product and assay based variations were also observed in the protamine neutralization profile of these LMWHs. Moreover, marked differences in some assays were observed when different batches of the generic copies of LMWHs were tested. Additional studies carried out to profile the oligosaccharide composition also showed product and batch-dependent variations. The relative amounts of antithrombin affinity components among the different generic products and within product batches also exhibited some variations. These studies clearly demonstrate that some of the generic copies of enoxaparin may not produce comparable anticoagulant and thrombin generation inhibitory effects at anti-Xa potency adjusted doses. Such differences may not be clinically relevant in the prophylactic indications (dosages ≤ 40 mg O.D). However, in therapeutic or interventional indications (IV or SC dosage &gt; 100 mg), these products may exhibit differential safety/efficacy profiles. These observations underscore the importance of clear guidelines on the chemical and biologic specifications for the acceptance of generic versions of LMWHs. Such measures are crucial to avoid any potential safety/inefficacy issues particulatly in indications where these drugs are used at higher dosages.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4385-4385
Author(s):  
Walter Jeske ◽  
Elizabeth McGeehan ◽  
Omer Iqbal ◽  
Debra Hoppensteadt ◽  
Jeanine M. Walenga ◽  
...  

Abstract Abstract 4385 Several biosimilar versions of branded enoxaparin (Lovenox, Sanofi-Aventis, Paris, France) have recently become available throughout the world. These biosimilar enoxaparin preparations are distributed by multiple suppliers in Asia and in North and South America. Enoxaparin represents a complex mixture of oligosaccharides obtained by alkaline depolymerization of porcine mucosal heparin. It is the most widely used low molecular weight heparin which has been validated for clinical use in multiple indications. While the molecular profile and anti-Xa potencies of some of the biosimilar versions of enoxaparin are comparable, product based differences have been reported amongst some of the biosimilar versions of enoxaparin. The purpose of this study was to compare the biochemical and pharmacologic profile of one biosimilar version of enoxaparin, namely Fibrinox (Sandoz SA, Buenos Aires, Argentina) with the branded product Lovenox. The products were compared in equigravimetric amounts, assuming equivalent potency (100 AXa U/mg). Both products exhibited comparable molecular weight profiles in terms of average molecular weight and oligosachharide distribution. Analysis of the antithrombin binding hexasaccharide fractions of Fibrinox and Lovenox indicated the presence of eight distinct hexasaccharides. The relative proportions these hexasaccharides differed between Fibrinox and Lovenox. The anti-Xa and anti-IIa activities were comparable. In the whole blood clot-based assays such as TEG and ACT, both agents produced similar anticoagulant effects. In the plasma based assays such as the APTT, Heptest and thrombin time, both products showed comparable anticoagulant effects in the normal human pooled plasma samples. However, in plasma samples collected from patients with liver disease who were apparently anticoagulant free, the two products showed differences in their anticoagulant effects in the APTT assay (p<0.05). In the TF mediated thrombin generation assay, Fibrinox produced a stronger inhibition of thrombin generation compared to Lovenox (IC50; Fibrinox, 1.6 μ g/ml, Lovenox 2.2 μ g/ml). No differences were observed between the two products in the agonist induced platelet aggregation assays. However in the 14C serotonin release study, Fibrinox produced a stronger HIT serum mediated 14C release (p<0.05). Differences in the fibrinokinetic profile and the inhibition of thrombin activatable fibrinolytic inhibitor activation were observed with these LMWHs. These studies suggest while both the molecular profile and the pharmacopoeial potency of Fibrinox is similar to the branded product, these drugs can be differentiated in some of the other assays and should be evaluated in terms of additional pharmacologic mechanisims to demonstrate bioequivalence. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2090-2090 ◽  
Author(s):  
Pierre Gueret ◽  
Chantal Krezel ◽  
Paul van Giersbergen ◽  
Eliane Fuseau ◽  
Thierry Lamy ◽  
...  

Abstract Abstract 2090 Poster Board II-67 EP42675 is the first representative of a new class of synthetic parenteral anticoagulants with a dual mechanism of action. EP42675 is an indirect factor Xa inhibitor (antithrombin binding pentasaccharide: fondaparinux analog) and a direct free and clot-bound thrombin (factor IIa) active site inhibitor (peptidomimetic: α-NAPAP analog). EP42675 was assessed in two phase I trials: (i) a randomized double-blind placebo-controlled single ascending dose study, where a single intravenous (IV) bolus of 1, 3, or 10 mg was administered to 3 groups of healthy male subjects (6 EP42675 and 2 matching placebo per group); (ii) a study assessing the effect of acetylsalicylic acid (ASA) plus clopidogrel, and unfractionated heparin (UFH) on pharmacokinetics (PK) and pharmacodynamics (PD) of EP42675. In this study, 24 healthy male subjects received 5 mg of EP42675 as an IV bolus on day 1, then ASA 100 mg and 75 mg clopidogrel from day 8 to 21. On day 15, they received a second IV bolus of 5 mg of EP42675 and were immediately randomized in 3 groups to receive either placebo, UFH 30 IU/Kg or UFH 60 IU/Kg, thus mimicking the rescue use of UFH in complicated percutaneous coronary interventions (PCI). EP42675 plasma concentrations were measured using anti-Xa and anti-IIa activity specific bioassays (Biomnis, France). EP42675 PD was assessed by global: thrombin generation test (TGT) and activated clotting time (ACT: ACT+ cartridge, Hemochron Signature Elite®, Gamida), and specific coagulation tests performed on a STA-R Evolution® automaton: prothrombin time (PT: Neoplastine CI plus Diagnostica Stago), activated partial thromboplastin time (aPTT: PTTA Diagnostica Stago), ecarin clotting time (ECT: Ecarine Diagnostica Stago), anti Xa activity expressed as ΔDO/min (Biophen Heparin Hyphen Biomed®), and thrombin time using purified human alpha thrombin (TT: Hemoclot Thrombin Inhibitors, Hyphen Biomed®). TGT performed on platelet poor plasma (PPP) were triggered with PPP reagent high (20 pM recombinant human tissue factor and 4 μM phospholipids, Biodis) on a Calibrated Automated Thrombogram (CAT™: Diagnostica Stago). The EP42675 plasma concentrations, measured by both anti-IIa and anti-Xa bioassays, showed a high and significant correlation (r=0.99, p<0.0001) indicating that the two active moieties of EP42675 did not dissociate. The plasma concentration versus time curves showed a marked distribution phase followed by a slow terminal phase and a less than proportional increase in exposure with increasing dose, with a half-life between 44.1 and 55.3 hours. EP42675 increased ACT, TGT lag time, TT, aPTT, PT, ECT, and anti-Xa activity. Maximum anticoagulant effect was reached within 2 minutes after bolus injection and lasted for more than 72 hours. The higher sensitivity of the TT test compared to the ECT test is explained by the higher selectivity of the EP42675 antithrombin moiety for the human alpha thrombin. A decrease in TGT endogenous thrombin potential was observed with a complete inhibition of thrombin generation at peak in the 10 mg dose group. The apparent lack of dose response at lower concentrations is likely to be due to interactions between the EP42675 antithrombin moiety with the alpha2 macroglobulin-thrombin complex. As anticipated, the co-administration of UFH induced a dose-dependent further increase in ACT, TGT lag time, TT, aPTT, PT, and anti-Xa activity, while the combination of ASA and clopidogrel did not influence the PK or PD of EP42675. The intra- and inter-subject variability was low for both PK and PD parameters. EP42675 was well tolerated. The only drug-related adverse events were mild hematoma at injection or blood sampling site in 6 subjects, mild gingival hemorrhage in 7 subjects, and mild epistaxis in one subject. No change in liver function tests was observed. These data provide useful information for designing future clinical studies with a single-dose anticoagulant treatment in patients with acute coronary syndrome undergoing PCI, and treated with various combinations of antithrombotic drugs. Disclosures: Gueret: Endotis: Research Funding. Krezel:Endotis: Employment. van Giersbergen:Endotis: Consultancy. Fuseau:Endotis: Consultancy. Neuhart:Endotis: Employment.


1979 ◽  
Author(s):  
R. Moncada ◽  
H. L. Messmore ◽  
J. Fareed ◽  
P. J. Scanlon ◽  
Z. Parvez

Although clinical incompatibilities of antihistamines and protamine with radiologic contrast media are well recognized, no report is available on the interaction of heparin, Coumadin, dextrans and other anticoagulants with these agents. We have employed the automated activated clotting time (ACT), prothrombin time (FT), partial thromboplastin time (PTT) and the thrombin time (TT) methods to monitor the anticoagulant actions of contrast media and its interaction with various anticoagulant drugs in patients undergoing angiography, A strong synergism of the anticoagulant action of heparin was observed in patients given heparin along with contrast media. Studies conducted in human volunteers revealed that contrast media at a 1-5 mg/ml level (clinical, 0.5-0.6 mg/ml) produce a strong synergistic effect on the anticoagulant action of heparin, oral anticoagulants, dextrans, and antiplatelet drugs. When blood obtained from patients undergoing angiography was supplemented with 0.25 u/ml heparin, the ACT, PTT and TT were equal to 1.5-2.0 units of heparin. Conventional amounts of protamine are incapable of neutralizing this synergistic interaction. These studies show that contrast media temporarily augments the degree of anticoagulation in patients undergoing angiography, which should be taken into consideration in patients undergoing vascular angiography.


Blood ◽  
2001 ◽  
Vol 97 (8) ◽  
pp. 2314-2322 ◽  
Author(s):  
Saulius Butenas ◽  
Kevin M. Cawthern ◽  
Cornelis van't Veer ◽  
Maria E. DiLorenzo ◽  
Jennifer B. Lock ◽  
...  

Abstract Several platelet inhibitors were examined in a tissue factor (TF)–initiated model of whole blood coagulation. In vitro coagulation of human blood from normal donors was initiated by 25 pM TF while contact pathway coagulation was suppressed using corn trypsin inhibitor. Products of the reaction were analyzed by immunoassay. Preactivation of platelets with the thrombin receptor activation peptide did not influence significantly the clotting time or thrombin–antithrombin III complex (TAT) formation. Addition of prostaglandin E1 (5 μM) caused a significant delay in clotting (10.0 minutes) versus control (4.3 minutes). The prolonged clotting time is correlated with delays in platelet activation, formation of TAT, and fibrinopeptide A (FPA) release. In blood from subjects receiving acetylsalicylic acid (ASA or aspirin), none of the measured products of coagulation were significantly affected. Similarly, no significant effect was observed when 5 μM dipyridamole (Persantine) was added to the blood. Antagonists of the platelet integrin receptor glycoprotein (gp) IIb/IIIa had intermediate effects on the reaction. A 1- to 2-minute delay in clot time and FPA formation was observed with addition of the antibodies 7E3 and Reopro (abciximab) (10 μg/mL), accompanied by a 40% to 70% reduction in the maximal rate of TAT formation and delay in platelet activation. The cyclic heptapetide, Integrilin (eptifibatide), at 5 μM concentration slightly prolonged clot time and significantly attenuated the maximum rate of TAT formation. The disruption of the gpIIb/IIIa-ligand interaction not only affects platelet aggregation, but also decreases the rate of TF-initiated thrombin generation in whole blood, demonstrating a potent antithrombotic effect superimposed on the antiaggregation characteristics.


Sign in / Sign up

Export Citation Format

Share Document