Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Br J Anaesth ; 121(4): 758-767, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30236238

ABSTRACT

BACKGROUND: We investigated potential for hypersensitivity reactions after repeated sugammadex administration and explored the mechanism of hypersensitivity. METHODS: In this double-blind, placebo-controlled study (NCT00988065), 448 healthy volunteers were randomised to one of three arms to receive three repeat i.v. administrations of either sugammadex 4 mg kg-1, 16 mg kg-1, or placebo. Primary endpoint was percentage of subjects with hypersensitivity (assessed by an independent adjudication committee). Secondary endpoint of anaphylaxis was classified per Sampson and Brighton criteria. Exploratory endpoints included skin testing, serum tryptase, anti-sugammadex antibodies [immunoglobulin (Ig) E/IgG], and other immunologic parameters. RESULTS: Hypersensitivity was adjudicated for 1/148 (0.7%), 7/150 (4.7%), and 0/150 (0.0%) subjects after sugammadex 4 mg kg-1, 16 mg kg-1, and placebo, respectively. After sugammadex 16 mg kg-1, one subject met Sampson criterion 1 and Brighton level 1 (highest certainty) anaphylaxis criteria; two met Brighton level 2 criteria. After database lock it was determined that certain protocol deviations could have introduced bias in the reporting of hypersensitivity signs/symptoms in a subject subset. Objective laboratory investigations indicated that potential underlying hypersensitivity mechanisms were unlikely to have been activated; the results suggest that most of the observed hypersensitivity reactions were unlikely IgE/IgG-mediated. CONCLUSION: Dose-dependent hypersensitivity or anaphylaxis reactions to sugammadex were observed when administered without prior neuromuscular blocking agent. Laboratory investigations do not suggest prevalent allergen-specific IgE/IgG-mediated immunologic hypersensitivity. Because it could not be fully excluded that estimates of hypersensitivity/anaphylaxis incidence were unbiased, an additional study was conducted to characterise the potential for hypersensitivity reactions and is described in a companion report. CLINICAL TRIAL REGISTRATION: http://www.clinicaltrials.gov NCT00988065; Protocol number P06042.


Subject(s)
Drug Hypersensitivity/immunology , Sugammadex/adverse effects , Administration, Intravenous , Adolescent , Adult , Anaphylaxis/immunology , Antibodies/immunology , Double-Blind Method , Female , Healthy Volunteers , Humans , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Male , Middle Aged , Safety , Skin Tests , Sugammadex/administration & dosage , Tryptases/blood , Young Adult
2.
Vox Sang ; 112(8): 704-712, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28952153

ABSTRACT

BACKGROUND AND OBJECTIVES: Platelet transfusions are widely administered to restore perioperative haemostasis in haemorrhagic patients; however, the role of platelet transfusion is not well understood and administration is often based on empiric data. This review aims to explore consensus regarding platelet transfusion trigger, dose and how the haemostatic efficacy of platelet transfusion was assessed for the treatment of perioperative bleeding. MATERIALS AND METHODS: A literature search was carried out using MEDLINE (PubMed) on 28 February 2017, to identify publications reporting the effect of platelet transfusion in relation to triggers, dose and assessment of haemostatic efficacy in bleeding patients in a perioperative setting. RESULTS: Eight publications were identified across a variety of settings, covering both prophylactic and therapeutic platelet transfusion in adult patients; the majority of the reports were in cardiac surgery. A high degree of variability was observed in the published studies, with only 50% of articles specifying a trigger for platelet transfusion. The most commonly used trigger was platelet count (25% of publications), with no consensus identified regarding the platelet count values used as triggers. Doses reported per transfusion varied from 1 to 12 units, and outcome measures were mixed, although the majority of publications (63%) assessed the requirement for transfusion with other blood products. CONCLUSION: The lack of consensus in published studies hinders our ability to draw conclusions regarding platelet transfusion and highlights the need for further studies to assess the appropriate dose and triggers for use in perioperative patients.


Subject(s)
Hemorrhage/therapy , Platelet Transfusion , Hemostasis, Surgical , Humans , Perioperative Care , Thrombocytopenia/prevention & control
3.
Vox Sang ; 112(5): 443-452, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28466601

ABSTRACT

BACKGROUND AND OBJECTIVES: Extracorporeal membrane oxygenation (ECMO) is a method of life support for either isolated cardiac failure or respiratory failure, with or without cardiac failure. When used for hemodynamic support, the ECMO circuit presents a non-endothelialized, artificial surface to blood inciting an inflammatory response which activates haemostatic pathways. Anticoagulation may complicate a pre-existing coagulopathy and/or inadequate surgical hemostasis of varying severity. There is no standardized method to achieve and monitor anticoagulation or guide transfusion therapy during ECMO. We tested the hypothesis that institutions across the world conduct similar management of anticoagulation and transfusion during adult ECMO support. METHODS: This is a descriptive, self-reporting cross-sectional survey of anticoagulation and transfusion practice for patients age 18 or older on ECMO. This 38 multiple-choice question survey was sent to 166 institutions, internationally, utilizing adult ECMO. About 32·4% (54) of institutions responded. Responses were anonymously collected. Descriptive analyses were calculated. RESULTS: Our findings indicate there appears to be a significant practice variation among institutions regarding anticoagulation and transfusion during adult ECMO support. DISCUSSION: The lack of standard practices among institutions may reflect a paucity of data regarding optimal anticoagulation and transfusion for patients requiring ECMO. Standardized protocols for anticoagulation and transfusion may help increase quality of care for and reduce morbidity, mortality and cost to patients and healthcare centres. Further study is required for standardized, high quality care.


Subject(s)
Blood Coagulation , Blood Transfusion/methods , Extracorporeal Membrane Oxygenation/methods , Anticoagulants/pharmacology , Cross-Sectional Studies , Health Care Surveys , Heparin/pharmacology , Humans , Whole Blood Coagulation Time
4.
Br J Anaesth ; 117(suppl 3): iii18-iii30, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27940453

ABSTRACT

Perioperative bleeding remains a major complication during and after surgery, resulting in increased morbidity and mortality. The principal causes of non-vascular sources of haemostatic perioperative bleeding are a preexisting undetected bleeding disorder, the nature of the operation itself, or acquired coagulation abnormalities secondary to haemorrhage, haemodilution, or haemostatic factor consumption. In the bleeding patient, standard therapeutic approaches include allogeneic blood product administration, concomitant pharmacologic agents, and increasing application of purified and recombinant haemostatic factors. Multiple haemostatic changes occur perioperatively after trauma and complex surgical procedures including cardiac surgery and liver transplantation. Novel strategies for both prophylaxis and therapy of perioperative bleeding include tranexamic acid, desmopressin, fibrinogen and prothrombin complex concentrates. Point-of-care patient testing using thromboelastography, rotational thromboelastometry, and platelet function assays has allowed for more detailed assessment of specific targeted therapy for haemostasis. Strategic multimodal management is needed to improve management, reduce allogeneic blood product administration, and minimize associated risks related to transfusion.


Subject(s)
Blood Loss, Surgical , Hemorrhage/therapy , Intraoperative Complications/therapy , Perioperative Care/methods , Postoperative Complications/therapy , Hemorrhage/drug therapy , Hemostatic Techniques , Hemostatics/therapeutic use , Humans , Intraoperative Complications/blood , Postoperative Complications/blood
5.
Br J Anaesth ; 117(1): 41-51, 2016 07.
Article in English | MEDLINE | ID: mdl-27317703

ABSTRACT

BACKGROUND: Single-dose human fibrinogen concentrate (FCH) might have haemostatic benefits in complex cardiovascular surgery. METHODS: Patients undergoing elective aortic surgery requiring cardiopulmonary bypass were randomly assigned to receive FCH or placebo. Study medication was administered to patients with a 5 min bleeding mass of 60-250 g after separation from bypass and surgical haemostasis. A standardized algorithm for allogeneic blood product transfusion was followed if bleeding continued after study medication. RESULTS: 519 patients from 34 centres were randomized, of whom 152 (29%) met inclusion criteria for study medication. Median (IQR) pretreatment 5 min bleeding mass was 107 (76-138) and 91 (71-112) g in the FCH and placebo groups, respectively (P=0.13). More allogeneic blood product units were administered during the first 24 h after FCH, 5.0 (2.0-11.0), when compared with placebo, 3.0 (0.0-7.0), P=0.026. Fewer patients avoided transfusion in the FCH group (15.4%) compared with placebo (28.4%), P=0.047. The FCH immediately increased plasma fibrinogen concentration and fibrin-based clot strength. Adverse event rates were comparable in each group. CONCLUSIONS: Human fibrinogen concentrate was associated with increased allogeneic blood product transfusion, an unexpected finding contrary to previous studies. Human fibrinogen concentrate may not be effective in this setting when administered according to 5-minute bleeding mass. Low bleeding rates and normal-range plasma fibrinogen concentrations before study medication, and variability in adherence to the complex transfusion algorithm, may have contributed to these results. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov identifier no. NCT01475669; EudraCT trial no. 2011-002685-20.


Subject(s)
Cardiopulmonary Bypass , Cardiovascular Surgical Procedures , Fibrinogen/therapeutic use , Hemorrhage/drug therapy , Hemostatics/therapeutic use , Adult , Aged , Aged, 80 and over , Blood Transfusion/statistics & numerical data , Double-Blind Method , Female , Hemostasis, Surgical , Humans , Male , Middle Aged , Young Adult
6.
Br J Anaesth ; 113(6): 922-34, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24972790

ABSTRACT

Cryoprecipitate, originally developed as a therapy for patients with antihaemophilic factor deficiency, or haemophilia A, has been in use for almost 50 yr. However, cryoprecipitate is no longer administered according to its original purpose, and is now most commonly used to replenish fibrinogen levels in patients with acquired coagulopathy, such as in clinical settings with haemorrhage including cardiac surgery, trauma, liver transplantation (LT), or obstetric haemorrhage. Cryoprecipitate is a pooled product that does not undergo pathogen inactivation, and its administration has been associated with a number of adverse events, particularly transmission of blood-borne pathogens and transfusion-related acute lung injury. As a result of these safety concerns, along with emerging availability of alternative fibrinogen preparations, cryoprecipitate has been withdrawn from use in a number of European countries. Compared with the plasma from which it is prepared, cryoprecipitate contains a high concentration of coagulation factor VIII, coagulation factor XIII, and fibrinogen. Cryoprecipitate is usually licensed by regulatory authorities for the treatment of hypofibrinogenaemia, and recommended for supplementation when plasma fibrinogen levels decrease below 1 g litre(-1); however, this threshold is empiric and is not based on solid clinical evidence. Consequently, there is uncertainty over the appropriate dosing and optimal administration of cryoprecipitate, with some guidelines from professional societies to guide clinical practice. Randomized, controlled trials are needed to determine the clinical efficacy of cryoprecipitate, compared with the efficacy of alternative preparations. These trials will allow the development of evidence-based guidelines in order to inform physicians and guide clinical practice.


Subject(s)
Blood Coagulation Disorders/drug therapy , Coagulants/therapeutic use , Factor VIII/therapeutic use , Fibrinogen/therapeutic use , Coagulants/adverse effects , Coagulants/economics , Drug Administration Schedule , Drug Approval , Drug Costs/statistics & numerical data , Drug Monitoring/methods , Factor VIII/adverse effects , Factor VIII/economics , Fibrinogen/adverse effects , Fibrinogen/economics , Humans , Practice Guidelines as Topic
8.
Haemophilia ; 16(3): 510-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20050927

ABSTRACT

Activated prothrombin complex concentrates (aPCC) and recombinant activated factor VIIa (rFVIIa) are two important therapies in haemophilia patients with inhibitors and improve clot stability. We hypothesized that potential differences in procoagulant and fibrinolytic actions of aPCC and rFVIIa may lie in the clot stability against fibrinolytic activation. We used thrombin generation, fluorescence detection and thromboelastometry in anti-factor IXa (FIXa) aptamer-treated whole blood (WB) and plasma to evaluate: (i) generation of thrombin and activated factor X (FXa) and (ii) viscoelastic properties of blood clots in the presence of tissue plasminogen activator (tPA) after addition of aPCC (0.4 U mL(-1)) or rFVIIa (60 nm). Peak thrombin generation increased from 85 +/- 19 nm in aptamer-treated plasma to 276 +/- 83 nm and 119 +/- 22 nm after addition of aPCC and rFVIIa respectively (P < 0.001). FXa activity increased within 20 min by 87 +/- 6% and by 660 +/- 97% after addition of aPCC and rFVIIa respectively (P < 0.001). TPA-induced lysis time increased from 458 +/- 378 s in aptamer-treated WB to 1597 +/- 366 s (P = 0.001) and 1132 +/- 214 s (P = 0.075), after addition of aPCC and rFVIIa respectively. In this haemophilia model using the anti-FIXa aptamer, the larger amount of thrombin was generated with aPCC compared with rFVIIa, while FXa generation was more rapidly increased in the presence of rFVIIa. Furthermore, clot formation in anti-FIXa aptamer-treated WB was less susceptible to tPA-induced fibrinolysis after adding aPCC compared with rFVIIa.


Subject(s)
Blood Coagulation Factors/therapeutic use , Blood Coagulation/drug effects , Factor IXa/antagonists & inhibitors , Factor VIIa/therapeutic use , Hemophilia A/drug therapy , Thrombin/metabolism , Antifibrinolytic Agents/pharmacology , Blood Coagulation Factor Inhibitors/pharmacology , Factor VIII/antagonists & inhibitors , Factor Xa/metabolism , Fibrinolysis/drug effects , Humans , Recombinant Proteins/therapeutic use
9.
Br J Anaesth ; 104(3): 318-25, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20133450

ABSTRACT

BACKGROUND: Fibrinolysis contributes to coagulopathy after major trauma and surgery. We hypothesized that progressive haemodilution is responsible, at least in part, for increased fibrinolytic tendency of blood clot. METHODS: The study was performed in two parts. First, whole blood (WB) samples collected from six healthy, consented volunteers were diluted in vitro with either saline or fresh-frozen plasma (FFP) to 40% and 15% of baseline. We quantified factor levels related to coagulation and fibrinolysis, and measured endogenous thrombin generation in undiluted control plasma samples and in samples diluted with saline or FFP. Additionally, thromboelastometry was used to assess susceptibility to fibrinolysis after adding tissue plasminogen activator in undiluted WB samples and in samples diluted with saline before and after substitution of fibrinogen or FFP. Secondly, as a model of in vivo haemodilution, we evaluated the same parameters before and after operation in nine consented patients undergoing off-pump coronary artery bypass surgery. RESULTS: The dilution with saline caused dose-dependent decreases in plasma levels of coagulation and antifibrinolytic factors, and in thrombin generation. In FFP-supplemented samples, factor levels and thrombin generation were maintained within normal ranges. Fibrinolytic tendency was significantly higher after haemodilution with saline independent of fibrinogen substitution compared with FFP. Similarly, increased tendency for fibrinolysis was also observed in the in vivo haemodilution. CONCLUSIONS: We demonstrated in vitro and in vivo that progressive haemodilution decreases endogenous antifibrinolytic proteins including alpha(2)-antiplasmin and thrombin-activatable fibrinolysis inhibitor, resulting in increased fibrinolytic tendency. Therefore, early fluid replacement therapy with FFP might be advantageous after massive haemorrhage.


Subject(s)
Fibrinolysis/physiology , Hemodilution/adverse effects , Plasma , Adult , Aged , Antifibrinolytic Agents/blood , Carboxypeptidase B2/blood , Coronary Artery Bypass, Off-Pump , Female , Fibrinolysis/drug effects , Hemostasis, Surgical/methods , Humans , In Vitro Techniques , Male , Middle Aged , Sodium Chloride/pharmacology , Thrombelastography/methods , Thrombin/biosynthesis , Tissue Plasminogen Activator/pharmacology , Young Adult , alpha-2-Antiplasmin/analysis
10.
Transfus Apher Sci ; 43(1): 107-16, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20655807

ABSTRACT

The question of whether storage of red blood cells (RBCs) alters their capacity to deliver oxygen and affects patient outcomes remains in a state of clinical equipoise. Studies of the changes which occur while RBCs are stored have led to several physiologically plausible hypotheses that these changes impair RBC function when the units are transfused. Although there is some evidence of this effect in vivo from animal model experiments, the results of several largely retrospective patient studies have not been consistent. Some studies have shown an association between worse clinical outcomes and transfusion of RBC which have been stored for longer periods of time, while others have found no effect. Three multicenter, randomized, controlled trials have been developed to address this important, but currently unanswered, question. Two clinical trials, one in low birth weight neonates and the other in intensive care unit patients, are enrolling subjects in Canada (the Age of Red Blood Cells in Premature Infants; the Age of Blood Study). The third trial, which is being developed in the United States, is the Red Cell Storage Duration Study (RECESS). This is a multicenter, randomized, controlled trial in which patients undergoing complex cardiac surgical procedures who are likely to require RBC transfusion will be randomized to receive RBC units stored for either 10 or fewer days or 21 or more days. Randomization will only occur if the blood bank has enough units of RBC of both storage times to meet the crossmatch request; hence, subjects randomized to the 21 day arm will receive RBC of the same storage time as they would have following standard inventory practice of "oldest units out first". The primary outcome is the change in the Multiple Organ Dysfunction Score (MODS), a composite measure of multiorgan dysfunction, by day 7. Secondary outcomes include the change in the MODS by day 28, all-cause mortality, and several composite and single measures of specific organ system function. The estimated total sample size required will be 1434 evaluable subjects (717 per arm).


Subject(s)
Blood Preservation/methods , Erythrocyte Transfusion , Erythrocytes/metabolism , Adolescent , Adult , Blood Preservation/adverse effects , Cardiac Surgical Procedures/methods , Female , Humans , Male , Oxygen/blood , Treatment Outcome , Young Adult
11.
Br J Anaesth ; 102(6): 793-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19420005

ABSTRACT

BACKGROUND: Replacement of fibrinogen is presumably the key step in managing dilutional coagulopathy. We performed an in vitro study hypothesizing that there is a minimal fibrinogen concentration in diluted whole blood above which the rate of clot formation approaches normal. METHODS: Blood samples from six healthy volunteers were diluted 1:5 v/v with saline keeping haematocrit at 24% using red cell concentrates. We measured coagulation factors and thrombin generation in plasma at baseline and after dilution. Thromboelastometry was used to evaluate (i) speed and quality of clot formation in diluted samples supplemented with fibrinogen 50-300 mg dl(-1) and (ii) clot resistance to fibrinolysis. Diluted and undiluted samples with no added fibrinogen served as controls. RESULTS: Coagulation parameters and platelets were reduced by 74-85% after dilution. Peak thrombin generation was reduced by 56%. Adding fibrinogen led to a concentration-dependent improvement of all thromboelastometric parameters. The half maximal effective concentration (EC50) for fibrinogen replacement in haemodiluted blood was calculated to be 125 mg dl(-1). Adding tissue plasminogen activator, 0.15 microg ml(-1), led to a decrease of clot firmness and lysis time. CONCLUSIONS: The target plasma concentration for fibrinogen replacement was predicted by these in vitro results to be greater than 200 mg dl(-1) as only these concentrations optimized the rate of clot formation. This concentration is twice the level suggested by the current transfusion guidelines. Although improved, clots were prone to fibrinolysis indicating that the efficacy of fibrinogen therapy may be influenced by co-existing fibrinolytic tendency occurring during dilutional coagulopathy.


Subject(s)
Fibrinogen/pharmacology , Hemodilution , Blood Coagulation/drug effects , Dose-Response Relationship, Drug , Fibrinogen/administration & dosage , Fibrinolysis , Hematocrit , Humans , In Vitro Techniques , Platelet Count , Thrombelastography , Thrombin/biosynthesis , Tissue Plasminogen Activator/pharmacology
12.
Acta Anaesthesiol Scand ; 53(3): 325-31, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19243318

ABSTRACT

INTRODUCTION: Serine proteases and their inhibitors play an important role in physiological homeostasis including neuronal activity, hemostasis, and wound healing. Tissue plasminogen activator (tPA) is involved in normal neuronal plasticity and memory formation but can also be neurotoxic. We hypothesized that the serine protease inhibitor aprotinin confers neuronal protection by inhibiting tPA activity. METHODS: Using cultured rat dopaminergic neuroblasts (N27 line), tPA-induced cytotoxicity was quantitated by an MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay and flow cytometry using propidium iodide DNA staining. The anti-apoptotic effects of aprotinin and other protease inhibitors were also evaluated using these systems. RESULTS: Treatment of cultured neuroblasts with tPA (10-20 microg/ml) caused a dose-dependent decrease in cell viability (71.3+/-2.4 at 10 microg/ml down to 52.7+/-2.5% at 20 microg/m tPA, 24-h treatment), which was potentiated in the absence of serum in the culture medium (59.5+/-6.3% at 10 microg/ml down to 47.9+/-4.7% at 20 microg/ml). Aprotinin was effective in ameliorating cell death when administered 30 min before tPA exposure as shown by increased cell viability (91.8+/-0.6% at tPA at 20 microg/ml), but this protection was significantly reduced when aprotinin was administered after tPA. The efficacy of aprotinin as a neuroprotectant was equivalent or superior to other direct tPA antagonist peptides Glu-Gly-Arg-chlormethylketone (EGRck) and Phe-Pro-Arg-chlormethylketone (FPRck) in this setting. CONCLUSION: These data suggest that one of the mechanisms of neuroprotection afforded by aprotinin may be inhibition of tPA-mediated neurotoxicity.


Subject(s)
Antifibrinolytic Agents/pharmacology , Cytoprotection/drug effects , Neurons/drug effects , Tissue Plasminogen Activator/toxicity , Animals , Cell Line , Flow Cytometry , Neurons/cytology , Rats
13.
J Thromb Haemost ; 17(2): 283-294, 2019 02.
Article in English | MEDLINE | ID: mdl-30582882

ABSTRACT

The vascular endothelial surface is coated by the glycocalyx, a ubiquitous gel-like layer composed of a membrane-binding domain that contains proteoglycans, glycosaminoglycan side-chains, and plasma proteins such as albumin and antithrombin. The endothelial glycocalyx plays a critical role in maintaining vascular homeostasis. However, this component is highly vulnerable to damage and is also difficult to examine. Recent advances in analytical techniques have enabled biochemical, visual and computational investigation of this vascular component. The glycocalyx modulates leukocyte-endothelial interactions, thrombus formation and other processes that lead to microcirculatory dysfunction and critical organ injury in sepsis. It also acts as a regulator of vascular permeability and contains mechanosensors as well as receptors for growth factors and anticoagulants. During the initial onset of sepsis, the glycocalyx is damaged and circulating levels of glycocalyx components, including syndecans, heparan sulfate and hyaluronic acid, can be measured and are reportedly useful as biomarkers for sepsis. Also, a new methodology using side-stream dark-field imaging is now clinically available for assessing the glycocalyx. Multiple factors including hypervolemia and hyperglycemia are toxic to the glycocalyx, and several agents have been proposed as therapeutic modalities, although no single treatment has been proven to be clinically effective. In this article, we review the derangement of the glycocalyx in sepsis. Despite the accumulated knowledge regarding the important roles of the glycocalyx, the relationship between derangement of the endothelial glycocalyx and severity of sepsis or disseminated intravascular coagulation has not been adequately elucidated and further work is needed.


Subject(s)
Endothelial Cells/metabolism , Glycocalyx/metabolism , Sepsis/metabolism , Animals , Blood Coagulation , Capillary Permeability , Cell Adhesion , Endothelial Cells/pathology , Glycocalyx/pathology , Humans , Sepsis/pathology , Sepsis/therapy , Signal Transduction
14.
J Thromb Haemost ; 16(2): 231-241, 2018 02.
Article in English | MEDLINE | ID: mdl-29193703

ABSTRACT

The inflammatory response and the activation of coagulation are two important responses in a host's defense against infection. These mechanisms do not work independently, but cooperate in a complex and synchronous manner. Recent research has also shed light on the critical role of thrombus formation, which prevents the dissemination of microorganisms. The cellular components of blood vessels, i.e. leukocytes, platelets, erythrocytes, and vascular endothelial cells, play significant roles in the development of thrombi in combination with activation of the coagulation system. In addition to the cellular components, alarmins such as histones and high-mobility group box 1, microparticles and secreted granule proteins are all important for clot formation. In this summary, we review the pathophysiology of sepsis-induced coagulopathy and the role of cellular components and critical factors released from damaged cells. In addition, we review important therapeutic approaches that have been developed, are under investigation and are currently available in certain countries, including antithrombin, recombinant thrombomodulin, anti-Toll-like receptor 4 therapy, anti-damage associated molecular pattern therapy, and hemoadsorption with a polymyxin B-immobilized fiber column.


Subject(s)
Blood Coagulation , Blood Platelets/metabolism , Cell Communication , Endothelial Cells/metabolism , Inflammation/metabolism , Neutrophils/metabolism , Sepsis/metabolism , Thrombosis/metabolism , Alarmins/immunology , Alarmins/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Anticoagulants/therapeutic use , Blood Coagulation/drug effects , Blood Platelets/drug effects , Blood Platelets/immunology , Cell Communication/drug effects , Endothelial Cells/drug effects , Endothelial Cells/immunology , Humans , Inflammation/blood , Inflammation/drug therapy , Inflammation/immunology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Neutrophils/drug effects , Neutrophils/immunology , Platelet Aggregation Inhibitors/therapeutic use , Sepsis/blood , Sepsis/drug therapy , Sepsis/immunology , Signal Transduction , Thrombosis/blood , Thrombosis/drug therapy , Thrombosis/immunology
15.
J Thromb Haemost ; 16(1): 54-64, 2018 01.
Article in English | MEDLINE | ID: mdl-29106076

ABSTRACT

Essentials Specific reversal agents for managing severe factor Xa inhibitor-associated bleeding are lacking. We assessed 4-factor-prothrombin complex concentrate (4F-PCC) and tranexamic acid (TXA). 4F-PCC, but not TXA, reduced the prothrombin time and increased endogenous thrombin potential. These agents may be viable options for reversal of therapeutic doses of rivaroxaban. SUMMARY: Background Oral activated factor X inhibitors such as rivaroxaban are widely used, but specific reversal agents are lacking. Although four-factor prothrombin complex concentrate (4F-PCC) and tranexamic acid (TXA) are sometimes used to manage serious bleeding, their efficacy is unknown. Prior studies in healthy subjects taking rivaroxaban revealed that 4F-PCC partially reverses the prolonged prothrombin time (PT), and fully restores the endogenous thrombin potential (ETP). The effect of TXA has not been evaluated. Methods In this double-blind, parallel-group study, 147 healthy volunteers given rivaroxaban 20 mg twice daily for 3 days were randomized after their morning dose on day 4 to receive intravenous 4F-PCC (50 IU kg-1 ), TXA (1.0 g), or saline. Standardized punch biopsies were performed at baseline and after 4F-PCC, TXA or saline administration. Reversal was assessed by measuring bleeding duration and bleeding volume at biopsy sites, and by determining the PT and ETP. Results As compared with saline, 4F-PCC partially reversed the PT and completely reversed the ETP, whereas TXA had no effect. Neither 4F-PCC nor TXA reduced bleeding duration or volume. All treatments were well tolerated, with no recorded adverse events. Conclusions Although 4F-PCC reduced the PT and increased the ETP in volunteers given supratherapeutic doses of rivaroxaban, neither 4F-PCC nor TXA influenced punch biopsy bleeding.


Subject(s)
Antidotes/administration & dosage , Antifibrinolytic Agents/administration & dosage , Blood Coagulation Factors/administration & dosage , Blood Coagulation/drug effects , Factor Xa Inhibitors/adverse effects , Hemorrhage/prevention & control , Rivaroxaban/adverse effects , Tranexamic Acid/administration & dosage , Adolescent , Adult , Antidotes/adverse effects , Antifibrinolytic Agents/adverse effects , Blood Coagulation Factors/adverse effects , Double-Blind Method , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/pharmacokinetics , Female , Healthy Volunteers , Hemorrhage/blood , Hemorrhage/chemically induced , Humans , Kansas , Male , Middle Aged , Pilot Projects , Prothrombin Time , Rivaroxaban/administration & dosage , Rivaroxaban/pharmacokinetics , Tranexamic Acid/adverse effects , Young Adult
16.
J Thromb Haemost ; 15(7): 1317-1321, 2017 07.
Article in English | MEDLINE | ID: mdl-28426914

ABSTRACT

Essentials Reversal of anticoagulant effects of dabigatran may occur despite application of idarucizumab. Monitoring of dabigatran level after antidote application is crucial to detect rebound. Repeated doses of idarucizumab may be necessary in cases of massive dabigatran accumulation. Combination of antidote application and renal replacement therapy may offer additional benefit. SUMMARY: Idarucizumab is a monoclonal antibody fragment designed for reversing the anticoagulant effects of dabigatran. Administration is recommended as two intravenous boluses of 2.5 g within 15 min of each other or as a single 5 g bolus. However, in certain situations a second dose of the drug could be necessary. We report the case of a 77-year-old man, treated with dabigatran for paroxysmal atrial fibrillation. He presented at our department with acute renal failure, concomitant massive dabigatran accumulation and subsequent acute gastrointestinal bleeding. Fifty minutes after the administration of idarucizumab, the dabigatran plasma concentration decreased from a peak of 1630 ng ml-1 to a level below the detection limit of 30 ng ml-1 and bleeding stopped. Eight hours after administration, the dabigatran plasma level started to increase up to 1560 ng ml-1 (96% of the maximum value obtained), accompanied by a further drop in hemoglobin. Concomitant hemodialysis and hemofiltration led to a continuous decrease in dabigatran plasma levels. However, sepsis and multiorgan failure ensued, which led to death. With this case report we raise the question of whether massive dabigatran accumulation requires repeated doses of idarucizumab, or alternatively, if the combination of antidote with hemodialysis/renal replacement therapy is advisable in order to remove circulating levels of dabigatran.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Anticoagulants/administration & dosage , Dabigatran/adverse effects , Acute Kidney Injury/chemically induced , Acute Kidney Injury/complications , Aged , Antithrombins/administration & dosage , Antithrombins/adverse effects , Atrial Fibrillation/complications , Atrial Fibrillation/drug therapy , Blood Coagulation/drug effects , Dabigatran/administration & dosage , Drug Administration Schedule , Fatal Outcome , Gastrointestinal Hemorrhage/etiology , Hemofiltration , Hemorrhage/drug therapy , Humans , Male , Renal Dialysis , Renal Replacement Therapy , Sepsis/complications
18.
J Thorac Cardiovasc Surg ; 130(1): 107-13, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15999048

ABSTRACT

OBJECTIVES: We sought to evaluate the efficacy of recombinant human antithrombin III for restoration of heparin responsiveness in heparin-resistant patients scheduled for cardiac surgery. METHODS: This was a multicenter, randomized, double-blind, placebo-controlled study in heparin-resistant patients undergoing elective cardiac surgery. Patients were considered heparin resistant if the activated clotting time was less than 480 seconds after 400 U/kg heparin. Fifty-two heparin-resistant patients were randomized into 2 cohorts. One cohort received a single bolus (75 U/kg) of recombinant human antithrombin III (n = 28), and the other, the placebo group (n = 24), received a normal saline bolus. If the activated clotting time remained less than 480 seconds, this was defined as treatment failure, and 2 units of fresh frozen plasma were transfused. Patients were monitored for adverse events during hospitalization. RESULTS: Six (21%) of the patients in the recombinant human antithrombin III group received fresh frozen plasma transfusions compared with 22 (92%) of the placebo-treated patients ( P < .001). Two units of fresh frozen plasma did not restore heparin responsiveness. There was no increased incidence of adverse events associated with recombinant human antithrombin III administration. Postoperative 24-hour chest tube bleeding was not different in the 2 groups. Surrogate measures of hemostatic activation suggested that there was less activation of the hemostatic system during cardiopulmonary bypass in the recombinant human antithrombin III group. CONCLUSION: Treatment with recombinant human antithrombin III in a dose of 75 U/kg is effective in restoring heparin responsiveness and promoting therapeutic anticoagulation for cardiopulmonary bypass in the majority of heparin-resistant patients. Two units of fresh frozen plasma were insufficient to restore heparin responsiveness. There was no apparent increase in bleeding associated with recombinant human antithrombin III.


Subject(s)
Antithrombin III/administration & dosage , Fibrinolytic Agents/therapeutic use , Heparin/therapeutic use , Adult , Aged , Blood Coagulation/physiology , Cardiopulmonary Bypass , Coronary Artery Bypass , Double-Blind Method , Drug Resistance , Hemostasis, Surgical , Humans , Middle Aged , Peptide Hydrolases/blood , Recombinant Proteins/therapeutic use , Whole Blood Coagulation Time
20.
Chest ; 105(1): 62-4, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8275786

ABSTRACT

Reports evaluating amrinone's effects in normal (intact) human subjects are complex and difficult to interpret because of the drug's diverse effects on myocardial function, resistance vessels, and vascular capacitance. In this study, 1.5 mg/kg of amrinone was administered as an intravenous bolus to cardiac surgical patients during constant flow cardiopulmonary bypass to determine its isolated effects on venous capacitance and vascular resistance. We noted a significant decrease in vascular resistance up to 8 min after injection and a 460 +/- 160 ml (mean +/- SD) decrease in venous reservoir volume 10 min after injection. Amrinone has potent venous and arterial vasodilating properties that make it a unique drug for treating congestive heart failure in addition to its reported positive inotropic effects.


Subject(s)
Amrinone/pharmacology , Vascular Resistance/drug effects , Vasodilation/drug effects , Arteries/drug effects , Blood Circulation/drug effects , Blood Pressure/drug effects , Blood Volume/drug effects , Cardiopulmonary Bypass , Heart Failure/drug therapy , Heart Failure/physiopathology , Humans , Veins/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL