Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 320
Filter
1.
Ann Card Anaesth ; 27(3): 202-212, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38963354

ABSTRACT

ABSTRACT: Protamine, first isolated from salmon fish sperm and now produced through recombinant biotechnology, is an antidote that neutralizes the anticoagulant properties of heparin. Protamine function is based on the capacity to dissociate the heparin-antithrombin III (AT III) complex (an important link that promotes blood fluidification by inhibiting coagulation), forming the inactive heparin-protamine complex. Protamine has itself dose-dependent anticoagulant properties: It interferes with coagulation factors and platelet function; it stimulates fibrinolysis; it can lead to thrombocytopenia and reduction in thrombin-related platelet aggregation; it decreases platelet response to thrombin receptor agonist in a dose-dependent manner. In this review, we will focus on protamine and its interaction with heparin. Notably, protamine is able to antagonize not only unfractionated heparin (UFH) but also low molecular weight heparins to various degrees. Protamine-allergic and anaphylactoid systemic reactions may affect up to 1 in 10 people and should be prevented and treated early.


Subject(s)
Anticoagulants , Heparin Antagonists , Heparin , Protamines , Humans , Heparin Antagonists/pharmacology , Heparin Antagonists/therapeutic use , Anticoagulants/pharmacology , Drug Interactions , Animals
2.
Clin Appl Thromb Hemost ; 30: 10760296241247558, 2024.
Article in English | MEDLINE | ID: mdl-38656136

ABSTRACT

INTRODUCTION: Andexanet alfa (AA) - zhzo, recombinant coagulation factor Xa, is an approved antidote for oral Xa inhibitors (apixaban and rivaroxaban). Unfractionated heparin (UFH) is commonly used for therapeutic, interventional, and surgical indications. Protamine sulfate (PrSO4) is frequently used to neutralize UFH. This study aimed to investigate the comparative neutralization profiles of AA and PrSO4 for heparins of bovine, ovine, and porcine origin. MATERIALS AND METHODS: The neutralization effect of PrSO4 at 25 µg/ml and AA at 100 µg/ml was studied on an approximate surgical/interventional concentration of heparin by supplementing whole blood with each of the heparins at 25 µg/ml. For the clotting profile (activated partial thromboplastin time: aPTT), amidolytic (anti-Xa and anti-IIa), and thrombin generation assay each of the heparin were supplemented from -10-0.62 µg/ml. RESULTS: In the whole blood ACT studies, all three heparins produced strong anti-coagulant effects (400-450 seconds) compared to saline (130-150 seconds). Both AA and PrSO4 almost fully neutralized the anti-coagulant effects of heparins (140-160 seconds). Both antidotes completely reversed the anticoagulant effects of all three heparins in the aPTT and thrombin generation assay. However, PrSO4 was more effective in neutralizing the anti-Xa, and anti-IIa effects than AA, which only partially neutralized these effects. CONCLUSION: Andexanet alfa at 100 µg/ml effectively neutralizes the therapeutic and surgical/interventional concentrations of heparins in in-vitro settings. While differences in the anti-Xa, and anti-IIa effects between heparins were noted, anti-coagulant effect of these agents in the aPTT assay were comparable. A similar neutralization profile was observed in the ACT and thrombin generation assays by both agents.


Subject(s)
Factor Xa , Heparin Antagonists , Heparin , Protamines , Recombinant Proteins , Recombinant Proteins/pharmacology , Factor Xa/pharmacology , Cattle , Sheep , Swine , Animals , Anticoagulants/pharmacology , Heparin/pharmacology , Protamines/pharmacology , Heparin Antagonists/pharmacology , Blood Coagulation/drug effects , Thrombin Time
3.
Perfusion ; 38(8): 1631-1636, 2023 11.
Article in English | MEDLINE | ID: mdl-36036659

ABSTRACT

INTRODUCTION: Our study aim was to explore how different protamine-heparin ratios impacted enzymatic coagulation and acellular fibrin clot growth in plasma using an in vitro model. We hypothesized that a low protamine-heparin ratio would be associated with superior fibrin clot growth dynamics. METHODS: We performed an in vitro study using 15 plasma samples from a commercial supplier. Different protamine-heparin ratios were added to each donor plasma sample: low ratio (0.7-1), traditional ratio (1-1), and high ratio (1.3-1) and clot formation dynamics were evaluated using a Thrombodynamics analyzer. Study outcomes were initial clot growth velocity and clot size at 30 min. RESULTS: Plasma samples treated with a one-to-one protamine-heparin ratio had significantly lower mean initial clot growth velocity compared to samples treated with a low protamine-heparin ratio; mean difference -2.3 µm/min (95% CI = -4.0 to -0.7, p = .004). Plasma samples treated with a one-to-one protamine-heparin ratio also had significantly smaller mean clot size at 30 min compared to samples treated with a low protamine-heparin ratio; mean difference -54.0 µm (95% CI = -107.6 to -0.4, p = .048). There were no significant differences in mean initial clot growth velocity or clot size at 30 min between plasma samples treated with a high protamine-heparin ratio and those treated with a one-to-one or low protamine-heparin ratio (all p > .05). CONCLUSIONS: Plasma samples treated with a low protamine-heparin ratio had superior clot growth velocity and larger clot size at 30 min compared to a one-to-one ratio, supporting the notion that a low protamine-heparin ratio may optimize enzymatic coagulation after cardiopulmonary bypass.


Subject(s)
Heparin , Protamines , Humans , Heparin/pharmacology , Protamines/pharmacology , Fibrin , Anticoagulants , Heparin Antagonists/pharmacology , Heparin Antagonists/therapeutic use , Cardiopulmonary Bypass
5.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34681808

ABSTRACT

Uncontrolled bleeding after enoxaparin (ENX) is rare but may be life-threatening. The only registered antidote for ENX, protamine sulfate (PS), has 60% efficacy and can cause severe adverse side effects. We developed a diblock copolymer, heparin-binding copolymer (HBC), that reverses intravenously administered heparins. Here, we focused on the HBC inhibitory activity against subcutaneously administered ENX in healthy mice. BALB/c mice were subcutaneously injected with ENX at the dose of 5 mg/kg. After 110 min, vehicle, HBC (6.25 and 12.5 mg/kg), or PS (5 and 10 mg/kg) were administered into the tail vein. The blood was collected after 3, 10, 60, 120, 360, and 600 min after vehicle, HBC, or PS administration. The activities of antifactors Xa and IIa and biochemical parameters were measured. The main organs were collected for histological analysis. HBC at the lower dose reversed the effect of ENX on antifactor Xa activity for 10 min after antidote administration, whereas at the higher dose, HBC reversed the effect on antifactor Xa activity throughout the course of the experiment. Both doses of HBC completely reversed the effect of ENX on antifactor IIa activity. PS did not reverse antifactor Xa activity and partially reversed antifactor IIa activity. HBC modulated biochemical parameters. Histopathological analysis showed changes in the liver, lungs, and spleen of mice treated with HBC and in the lungs and heart of mice treated with PS. HBC administered in an appropriate dose might be an efficient substitute for PS to reverse significantly increased anticoagulant activity that may be connected with major bleeding in patients receiving ENX subcutaneously.


Subject(s)
Enoxaparin/adverse effects , Hemorrhage/drug therapy , Protamines/therapeutic use , Animals , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Blood Coagulation Tests , Drug-Related Side Effects and Adverse Reactions/drug therapy , Drug-Related Side Effects and Adverse Reactions/prevention & control , Enoxaparin/administration & dosage , Female , Hemorrhage/chemically induced , Hemorrhage/prevention & control , Heparin/metabolism , Heparin Antagonists/metabolism , Heparin Antagonists/pharmacology , Heparin Antagonists/therapeutic use , Infusions, Subcutaneous , Male , Mice , Mice, Inbred BALB C , Polymers/chemistry , Polymers/metabolism , Polymers/pharmacology , Polymers/therapeutic use , Protamines/metabolism , Protamines/pharmacology , Protein Binding
7.
FEBS Open Bio ; 11(9): 2468-2489, 2021 09.
Article in English | MEDLINE | ID: mdl-34184429

ABSTRACT

Unfractionated heparin (UFH) is an anionic glycosaminoglycan that is widely used to prevent blood clotting. However, in certain cases, unwanted side effects can require it to be neutralized. Protamine sulfate (PS), a basic peptide rich in arginine, is the only approved antagonist for UFH neutralization. Many adverse reactions occur with the clinical application of PS, including systemic hypotension, pulmonary hypertension, and anaphylaxis. We previously described R15, a linear peptide composed of 15 arginine molecules, as a potential UFH antagonist. In this study, the in-depth safety of R15 was explored to reveal its merits and associated risks in comparison with PS. In vitro safety studies investigated the interactions of R15 with erythrocytes, fibrin, complement, and rat plasma. In vivo safety studies explored potential toxicity and immunogenicity of R15 and the UFH-R15 complex. Results showed that both PS and R15 can induce erythrocyte aggregation, thicken fibrin fibers, activate complement, and cause anticoagulation in a concentration-dependent manner. However, those influences weakened in whole blood or in live animals and were avoided when R15 was in a complex with UFH. We found dramatically enhanced complement activation when there was excess UFH in analyses involving UFH-PS complexes, and a slight increase in those involving UFH-R15 complexes. Within 2 h, R15 was degraded in rat plasma in vitro, whereas PS was not. Enhanced creatinine was found after a single intravenous injection of PS or R15 (900 U·kg-1 , body weight), suggesting possible abnormal renal function. The UFH-PS complex, but not the UFH-R15 complex, exhibited obvious immunogenicity. In conclusion, R15 is nonimmunogenic and potentially safe at a therapeutic dose to reverse the effects of UFH.


Subject(s)
Heparin Antagonists/pharmacology , Heparin/pharmacology , Peptides/pharmacology , Animals , Blood Coagulation/drug effects , Blood Coagulation Tests , Complement Activation , Drug Antagonism , Erythrocytes/drug effects , Erythrocytes/metabolism , Fibrin/metabolism , Heparin Antagonists/chemistry , Humans , Peptides/chemistry , Protamines/chemistry , Protamines/pharmacology , Rats
8.
Ann Pharmacother ; 55(2): 261-264, 2021 02.
Article in English | MEDLINE | ID: mdl-32667214

ABSTRACT

The recent shortage of protamine prompted an investigation of alternatives for reversal of unfractionated heparin. Heparin is an anticoagulant utilized in the hospital setting. Available options for anticoagulation include direct oral anticoagulants, vitamin K antagonists, thrombin inhibitors, low-molecular-weight heparins, and heparin. Protamine is the approved reversal agent for heparin with few alternatives under investigation. Although andexanet was designed as an antidote for apixaban and rivaroxaban, in vitro studies show that in a dose-dependent technique, andexanet had near full reversal of heparin, reversed anti-factor Xa activity, and neutralized anticoagulant effects of activated partial thromboplastin time and thrombin time induced by heparin.


Subject(s)
Blood Coagulation/drug effects , Factor Xa/therapeutic use , Heparin Antagonists/therapeutic use , Protamines/therapeutic use , Recombinant Proteins/therapeutic use , Anticoagulants/adverse effects , Blood Coagulation Tests , Factor Xa/administration & dosage , Factor Xa/adverse effects , Hemorrhage/chemically induced , Hemorrhage/prevention & control , Heparin/adverse effects , Heparin Antagonists/administration & dosage , Heparin Antagonists/adverse effects , Heparin Antagonists/pharmacology , Humans , Protamines/administration & dosage , Protamines/adverse effects , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects
9.
Asian Cardiovasc Thorac Ann ; 29(1): 5-9, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32854516

ABSTRACT

BACKGROUND: Heparin is used for anticoagulation during cardiopulmonary bypass. After weaning from bypass, protamine is administered to neutralize the effects of heparin and thus reestablish hemostasis. Rotational thrombelastometry has been shown to discriminate between heparin and other impairing effects on coagulation. We analyzed the interaction of heparin and protamine under different conditions of overdosage in an in-vitro trial. METHODS: Blood samples were taken from 17 healthy volunteers, separated, and spiked in vitro with heparin, protamine for heparin neutralization, an overdosage of protamine, and two dosages of re-heparinization to evaluate heparin effects under the condition of protamine overdosage. All samples were analyzed in a standard ROTEM rotational thromboelastometry device after intrinsic activation with and without addition of heparinase. Coagulation time, maximum clot firmness, and clot formation time were recorded. RESULTS: Heparin led to prolongation of coagulation and clot formation times in the test without heparinase. Adequate protamine addition normalized the test, and overdosage of protamine led to significant prolongation of both times. Addition of heparin in the presence of protamine overdosage normalized these parameters. CONCLUSION: We reconfirmed that the ROTEM device enables discrimination of the effects heparin and protamine on coagulation and detection of the coagulation-impairing effects of protamine overdosage. Furthermore, we were able to show a positive effect on coagulation times by heparin in the presence of protamine overdosage. Because this was an in-vitro study, these findings need to be confirmed in vivo, requiring further research.


Subject(s)
Anticoagulants/pharmacology , Blood Coagulation/drug effects , Drug Overdose/blood , Heparin Antagonists/pharmacology , Heparin/pharmacology , Protamines/pharmacology , Adult , Female , Humans , Male , Thrombelastography , Time Factors
10.
Interact Cardiovasc Thorac Surg ; 31(3): 391-397, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32620960

ABSTRACT

OBJECTIVES: In cardiac surgery, adequate heparinization is necessary to prevent thrombus formation in the cardiopulmonary bypass (CPB). To counteract the heparin effect after weaning from CPB, protamine is administered. The optimal protamine/heparin ratio is still unknown. METHODS: In this before-after study, we evaluated the effect of a 0.6/1-protamine/heparin ratio implementation as of May 2017 versus a 0.8/1-protamine/heparin ratio on the 12-h postoperative blood loss and the amount of blood and blood component transfusions (fresh frozen plasma, packed red blood cells, fibrinogen concentrate, platelet concentrate and prothrombin complex concentrate) after cardiac surgery. A total of 2051 patients who underwent cardiac surgery requiring CPB between May 2016 and May 2018 were included. RESULTS: In the 0.6/1-protamine/heparin ratio group, only 28.8% of the patients received blood component transfusion, compared to 37.9% of the patients in the 0.8/1-ratio group (P < 0.001). The median 12-h postoperative blood loss was 230 ml (interquartile range 140-320) in the 0.6/1-ratio group versus 260 ml (interquartile range 155-365) in the 0.8/1-ratio group (P < 0.001). CONCLUSIONS: A 0.6/1-protamine/heparin ratio after weaning from CPB is associated with a significantly reduced 12-h postoperative blood loss and blood components transfusion.


Subject(s)
Blood Component Transfusion/trends , Cardiac Surgical Procedures , Heparin/pharmacology , Protamines/pharmacology , Aged , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Blood Loss, Surgical/prevention & control , Female , Heparin Antagonists/pharmacology , Humans , Male , Postoperative Hemorrhage/prevention & control
11.
Clin Appl Thromb Hemost ; 26: 1076029619895120, 2020.
Article in English | MEDLINE | ID: mdl-31914798

ABSTRACT

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.


Subject(s)
Factor Xa/pharmacology , Heparin Antagonists/pharmacology , Recombinant Proteins/pharmacology , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Blood Coagulation Tests , Drug Interactions , Enoxaparin/antagonists & inhibitors , Fondaparinux/antagonists & inhibitors , Humans , Thrombelastography
12.
J Thromb Haemost ; 16(9): 1789-1799, 2018 09.
Article in English | MEDLINE | ID: mdl-29968421

ABSTRACT

Essentials Bovine (HBI) and porcine (HPI) heparins differ in structure and anticoagulant activity. Protamine-neutralization was evaluated on a variety of physical-chemical methods. HBI requires more protamine than HPI to fully neutralize its anticoagulant activity. Protamine preferentially removes higher-sulfated chains of HBI while HPI is evenly precipitated. SUMMARY: Background Protamine neutralization is an essential step for the safe use and inactivation of the unfractionated heparin (UFH) that is widely employed in surgical and non-surgical procedures involving extracorporeal circulation. Objective To compare protamine neutralization of different pharmaceutical-grade UFHs prepared from porcine or bovine intestine (HPI and HBI, respectively). HBI has approximately half the anticoagulant potency of HPI, mostly as consequence of its fraction enriched with N-sulfated α-glucosamine disaccharides. Methods Protamine neutralization of HPI and HBI was evaluated with in vitro, ex vivo and in vivo assays. We also performed in-depth assessments of the complexation of protamine with these distinct UFHs by using nuclear magnetic resonance and mass spectroscopy. Results HPI and HBI interact similarly with protamine on a mass/mass basis; however, HBI requires more protamine than HPI to have its anticoagulant activity fully neutralized, because of its lower potency, which entails the use of higher doses. Nuclear magnetic resonance spectra revealed that HPI precipitates homogeneously with protamine. On the other hand, the low-sulfated fraction of HBI, enriched with N-sulfated α-glucosamine, precipitates at higher concentrations of protamine than the fraction more like HPI, with a preponderance of N,6-disulfated α-glucosamine disaccharides. Finally, mass spectroscopy spectra showed that some of the different peptide components of protamine interact preferentially with the heparins, irrespective of their animal origin. Conclusion Our results have important medical implications, indicating that protamine neutralization of HBI, determined exclusively by point-of-care coagulation assessments, must fail because of its lower-sulfated fraction with reduced anticoagulant activity that could remain in the circulation after the neutralization procedure.


Subject(s)
Anticoagulants/pharmacology , Heparin Antagonists/pharmacology , Heparin/pharmacology , Protamines/pharmacology , Animals , Anticoagulants/chemistry , Anticoagulants/isolation & purification , Biological Assay , Cattle , Chemical Precipitation , Chromatography, Affinity , Disaccharides/chemistry , Dose-Response Relationship, Drug , Heparin/chemistry , Heparin/isolation & purification , Intestinal Mucosa/chemistry , Mass Spectrometry , Nuclear Magnetic Resonance, Biomolecular , Partial Thromboplastin Time , Protamines/chemistry , Rats , Species Specificity , Sulfur/analysis , Swine
13.
Br J Anaesth ; 120(5): 914-927, 2018 May.
Article in English | MEDLINE | ID: mdl-29661409

ABSTRACT

Neutralisation of systemic anticoagulation with heparin in cardiac surgery with cardiopulmonary bypass requires protamine administration. If adequately dosed, protamine neutralises heparin and reduces the risk of postoperative bleeding. However, as its anticoagulant properties are particularly exerted in the absence of heparin, overdosing of protamine may contribute to bleeding and increased transfusion requirements. This narrative review describes the mechanisms underlying the anticoagulant properties and side-effects of protamine, and the impact of protamine dosing on the activated clotting time and point-of-care viscoelastic test results, and explains the distinct protamine dosing strategies in relation to haemostatic activation and postoperative bleeding. The available evidence suggests that protamine dosing should not exceed a protamine-to-heparin ratio of 1:1. In particular, protamine-to-heparin dosing ratios >1 are associated with more postoperative 12 h blood loss. The optimal protamine-to-heparin ratio in cardiac surgery has, however, not yet been elaborated, and may vary between 0.6 and 1.0 based on the initial heparin dose.


Subject(s)
Anticoagulants/pharmacology , Cardiopulmonary Bypass , Heparin Antagonists/pharmacology , Postoperative Hemorrhage/prevention & control , Protamines/pharmacology , Anticoagulants/adverse effects , Cardiac Surgical Procedures , Dose-Response Relationship, Drug , Heparin/administration & dosage , Heparin Antagonists/adverse effects , Humans , Protamines/adverse effects
14.
Eur J Pharmacol ; 827: 167-172, 2018 May 15.
Article in English | MEDLINE | ID: mdl-29555505

ABSTRACT

Etamsylate is indicated for several anti-hemorrhagic indications in human and veterinary medicine. However, etamsylate has been shown to be effective only in specific hemorrhagic situations. Furthermore, mechanism of action of etamsylate is not known but recent research has shown its ability to inhibit heparin binding to several growth factors. We have evaluated the ability of etamsylate to interfere with the activities of heparin. Effects of etamsylate on vasodilatory activity of heparin were evaluated in rat aortic segments. Influence of etamsylate on anticoagulant activity of heparin was evaluated in vitro by determining prothrombin (PT) time and activated partial thromboplastin time (aPTT) in dog blood and in vivo by determining the interference of systemic and topical etamsylate on heparin-induced extension in bleeding time (BT) in rats. Despite failing to inhibit heparin-induced vasodilation of rat aorta, etamsylate significantly reduced the increase in aPTT caused by heparin (+30.4 ±â€¯6.7% vs. +15.0 ±â€¯2.8% for etamsylate at 100 µM, P < 0.05). Etamsylate also antagonized the anticoagulant effects driven by heparin in vivo since prevented the heparin-induced increase in BT when systemically (i.p.) administered (+94.6 ±â€¯7.5% vs. +57.9 ±â€¯9.2% at 10 mg/kg, P < 0.05, vs. +22.2 ±â€¯16.8% at 30 mg/kg, P < 0.01). Additionally, topically applied etamsylate (125 mg/ml) significantly reduced heparin-induced BT increase (+102.5 ±â€¯3.2% vs. +54.0 ±â€¯5.8%, P < 0.01). These evidences show a pharmacological interference by etamsylate on heparin activities antagonizing pro-hemorrhagic effects of heparin in vitro and in vivo without inhibiting its vasodilatory properties. This ability could help to explain pharmacological effects of etamsylate and proposes its role for reversing pro-hemorrhagic states.


Subject(s)
Anticoagulants/pharmacology , Drug Antagonism , Ethamsylate/pharmacology , Hemostatics/pharmacology , Heparin Antagonists/pharmacology , Heparin/pharmacology , Animals , Dogs , Female , Male , Rats, Sprague-Dawley , Vasodilation/drug effects
15.
Br J Haematol ; 180(5): 715-720, 2018 03.
Article in English | MEDLINE | ID: mdl-29363751

ABSTRACT

Heparin anticoagulation followed by protamine reversal is commonly used in cardiopulmonary bypass (CPB). As an alternative to protamine, a recombinant inactive antithrombin (riAT) was designed as an antidote to heparin and was previously shown to be as potent as protamine in-vitro. In the present study, riAT was assessed for its ability to neutralize heparin after CPB in a rat model. After 60 min of CPB under heparin, rats received 5 mg/kg protamine, 37.5 mg/kg riAT or phosphate buffered saline (PBS) as placebo. Residual anticoagulant activity was assessed using the activated partial thromboplastin time assay before, and 10-30 min after reversion. Haemodynamic monitoring was performed and plasma histamine concentration was also measured. In this model, riAT appeared to be as efficient as protamine in neutralizing heparin. Ten minutes after injection, riAT and protamine both decreased heparin activity, to 1.8 ± 1.3 and 4.5 ± 1.4 u/ml, respectively (23.1 ± 5.1 u/ml in placebo group). Furthermore, evolution of mean carotid arterial pressure, heart rate and plasma histamine levels was comparable in rats treated with PBS or riAT, while protamine exhibited haemodynamic side effects and increased histamine plasma concentration. Thus, riAT could represent an advantage over protamine in CPB because it efficiently reverses heparin activity without negative effects on haemodynamic parameters and plasma histamine level.


Subject(s)
Anticoagulants/pharmacology , Cardiopulmonary Bypass , Heparin Antagonists/pharmacology , Heparin/pharmacology , Protamines/pharmacology , Animals , Antithrombins/pharmacology , Hemodynamics/drug effects , Histamine/metabolism , Male , Rats, Wistar
16.
Perfusion ; 32(6): 481-488, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28820026

ABSTRACT

INTRODUCTION: This manuscript represents a pilot study assessing the feasibility of a single-compartment, individualised, pharmacokinetic algorithm for protamine dosing after cardiopulmonary bypass. METHODS: A pilot cohort study in a specialist NHS cardiothoracic hospital targeting patients undergoing elective cardiac surgery using cardiopulmonary bypass. Patients received protamine doses according to a pharmacokinetic algorithm (n = 30) or using an empirical, fixed-dose model (n = 30). Categorical differences between the groups were evaluated using the Chi-squared test or Fisher's exact test. Continuous data was analysed using a paired Student's t-test for parametric data and the paired samples Wilcoxon test for non-parametric data. RESULTS: Patients who had protamine dosing according to the algorithm demonstrated a lower protamine requirement post-bypass relative to empirical management as measured by absolute dose (243 ± 49mg vs. 305 ± 34.7mg; p<0.001) and the heparin to protamine ratio (0.79 ± 0.12 vs. 1.1 ± 0.15; p<0.001). There was no difference in the pre- to post-bypass activated clotting time (ACT) ratio (1.05 ± 0.12 vs. 1.02 ± 0.15; p=0.9). Patients who received protamine according to the algorithm had no significant difference in transfusion requirement (13.3% vs. 30.0%; p=0.21). CONCLUSIONS: This study showed that an individualized pharmacokinetic algorithm for the reversal of heparin after cardiopulmonary bypass is feasible in comparison with a fixed dosing strategy and may reduce the protamine requirement following on-pump cardiac surgery.


Subject(s)
Cardiac Surgical Procedures/methods , Cardiopulmonary Bypass/methods , Heparin Antagonists/therapeutic use , Heparin/therapeutic use , Protamines/therapeutic use , Aged , Female , Heparin/pharmacology , Heparin Antagonists/pharmacology , Humans , Male , Pilot Projects , Protamines/pharmacokinetics
17.
Blood ; 129(10): 1368-1379, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28034889

ABSTRACT

Anticoagulant therapy-associated bleeding and pathological thrombosis pose serious risks to hospitalized patients. Both complications could be mitigated by developing new therapeutics that safely neutralize anticoagulant activity and inhibit activators of the intrinsic blood clotting pathway, such as polyphosphate (polyP) and extracellular nucleic acids. The latter strategy could reduce the use of anticoagulants, potentially decreasing bleeding events. However, previously described cationic inhibitors of polyP and extracellular nucleic acids exhibit both nonspecific binding and adverse effects on blood clotting that limit their use. Indeed, the polycation used to counteract heparin-associated bleeding in surgical settings, protamine, exhibits adverse effects. To address these clinical shortcomings, we developed a synthetic polycation, Universal Heparin Reversal Agent (UHRA), which is nontoxic and can neutralize the anticoagulant activity of heparins and the prothrombotic activity of polyP. Sharply contrasting protamine, we show that UHRA does not interact with fibrinogen, affect fibrin polymerization during clot formation, or abrogate plasma clotting. Using scanning electron microscopy, confocal microscopy, and clot lysis assays, we confirm that UHRA does not incorporate into clots, and that clots are stable with normal fibrin morphology. Conversely, protamine binds to the fibrin clot, which could explain how protamine instigates clot lysis and increases bleeding after surgery. Finally, studies in mice reveal that UHRA reverses heparin anticoagulant activity without the lung injury seen with protamine. The data presented here illustrate that UHRA could be safely used as an antidote during adverse therapeutic modulation of hemostasis.


Subject(s)
Antidotes/pharmacology , Blood Coagulation/drug effects , Hemorrhage/drug therapy , Heparin Antagonists/pharmacology , Animals , Anticoagulants/adverse effects , Hemorrhage/chemically induced , Heparin/adverse effects , Humans , Lung/drug effects , Mice , Polyamines , Polyelectrolytes , Protamines/adverse effects
18.
Optom Vis Sci ; 93(11): 1349-1355, 2016 11.
Article in English | MEDLINE | ID: mdl-27536976

ABSTRACT

PURPOSE: To investigate the ability of protamine, alone or in combination with other antimicrobial agents, to kill bacteria and fungi associated with contact lens-related keratitis. METHODS: The International Organization for Standardization 14729:2001 procedure was used to test the antimicrobial activity of solutions of protamine (23-228 µM) with and without polyhexamethylene biguanide (PHMB) and ethylenediamine tetra-acetic acid (EDTA). The recommended ISO panel of microbes along with six clinical isolates was tested. The effect of increasing sodium chloride concentration on the antimicrobial activity was also assessed. The cytotoxicity of the final protamine/EDTA/PHMB solution was measured using ISO 10993-5 standard assays. RESULTS: Protamine gave a dose-dependent antimicrobial effect, with the highest effect for most strains being at 228 µM (≥6 log reductions of viable bacteria and ≥1 log reduction of viable fungi). Addition of EDTA and PHMB increased the antimicrobial effect for all strains except Pseudomonas aeruginosa ATCC6538, which had optimum activity (≥6 log inhibition) even in protamine alone. The optimum antimicrobial activity of all microbes was achieved in 0.2% sodium chloride, but even in 0.8% sodium chloride, the activity met or exceeded the ISO standard (>3 log reductions for bacteria and >1 log reduction for fungi). None of the formulations was cytotoxic to mammalian cells. CONCLUSIONS: This study highlights the potential for protamine to be used for the development of effective multipurpose disinfection solutions. Further investigations such as stability, compatibility with contact lenses, and in vivo toxicity are warranted.


Subject(s)
Anti-Infective Agents/pharmacology , Bacteria/drug effects , Contact Lenses/microbiology , Disinfectants/pharmacology , Fungi/drug effects , Protamines/pharmacology , Bacteria/isolation & purification , Biguanides/pharmacology , Colony Count, Microbial , Contact Lens Solutions/pharmacology , Disinfection/methods , Drug Combinations , Fungi/isolation & purification , Heparin Antagonists/pharmacology , Humans
19.
Thromb Res ; 138: 121-129, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26709038

ABSTRACT

BACKGROUND: The global supply of unfractionated heparin (UFH) and all commercially available low molecular weight heparins (LMWH) remain dependent on animal sources, such as porcine intestine or bovine lung. Recent experience has shown that contamination of the supply chain (with over-sulfated chondroitin sulfates) can result in lethal toxicity. Fondaparinux is currently the only commercially available synthetic analog of heparin. We recently described a new class of chemoenzymatically synthesized heparin analogs. One of these compounds (S12-mer) is a dodecasaccharide consisting of an antithrombin-binding moiety with repeating units of IdoA2S-GlcNS6S and two 3-O-sulfate groups that confer the ability to bind protamine. OBJECTIVE/METHODS: We sought to further characterize this new compound in vitro using biochemical and global coagulation assays and in vivo using thrombosis and hemostasis assays. RESULTS: The anticoagulant activities of the Super 12-mer (S12-mer) and Enoxaparin in anti-factor Xa and plasma-based thrombin generation assays were roughly equivalent with a 50% reduction in peak thrombin generation occurring at approximately 325nM. When protamine was titrated against a fixed concentration of S12-mer in plasma or blood, the S12-mer displayed a significant restitution of thrombin generation and clot formation. In vivo, S12-mer inhibited venous thrombosis to a similar extent as Enoxaparin, with similar bleeding profiles. CONCLUSIONS: These data show that the S12-mer has almost identical efficacy to Enoxaparin in terms of FXa inhibition, while displaying significant reversibility with protamine. Taken together with the ability to ensure purity and homogeneity from batch to batch, the S12-mer is a promising new synthetic heparin analog with a potentially enhanced safety profile.


Subject(s)
Blood Coagulation/drug effects , Factor Xa Inhibitors/chemistry , Factor Xa Inhibitors/pharmacology , Heparin/analogs & derivatives , Heparin/pharmacology , Animals , Anticoagulants/pharmacology , Enoxaparin/pharmacology , Factor Xa/metabolism , Heparin Antagonists/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Protamines/pharmacology , Thrombin/metabolism
20.
Prikl Biokhim Mikrobiol ; 52(4): 421-8, 2016.
Article in English | MEDLINE | ID: mdl-29512988

ABSTRACT

Alkylated derivatives of low molecular weight chitosan with different substitution degrees of 98, 40, and 9% (I, II, and III respectively) have been synthesized. The structure of the obtained derivatives was defined by spectral assays (IR-spectroscopy and proton magnetic resonance). Chitosan derivatives were characterized with positive zeta-potential (33­51 mV) and solubility from 2 to 100 mg/mL in pH 7.4 and 25°C. It was shown that, at a concentration of 0.0014­0.0029 mg/mL, derivative I, as well as protamine sulfate, could be used to neutralize the anticoagulant activity of unfractionated or low molecular weight heparin. At a concentration of 0.0029­0.58 mg/mL, derivative I enhanced platelet aggregation, which would be necessary when hemostatic compounds or materials were used. Derivatives II and III enhanced platelet aggregation to a lesser extent.


Subject(s)
Anticoagulants , Blood Platelets/metabolism , Chitosan , Heparin Antagonists , Heparin , Platelet Aggregation/drug effects , Anticoagulants/chemistry , Anticoagulants/pharmacology , Chitosan/analogs & derivatives , Chitosan/chemistry , Chitosan/pharmacology , Heparin/chemistry , Heparin/pharmacology , Heparin Antagonists/chemistry , Heparin Antagonists/pharmacology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...